EP3308641B1 - Immunodeficient, genetically modified rodent and methods of use thereof - Google Patents

Immunodeficient, genetically modified rodent and methods of use thereof Download PDF

Info

Publication number
EP3308641B1
EP3308641B1 EP17184813.8A EP17184813A EP3308641B1 EP 3308641 B1 EP3308641 B1 EP 3308641B1 EP 17184813 A EP17184813 A EP 17184813A EP 3308641 B1 EP3308641 B1 EP 3308641B1
Authority
EP
European Patent Office
Prior art keywords
human
cells
rodent
promoter
genetically modified
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP17184813.8A
Other languages
German (de)
English (en)
French (fr)
Other versions
EP3308641A1 (en
Inventor
Richard Flavell
Till Strowig
Markus G. Manz
Chiara BORSOTTI
Madhav Dhodapkar
Andrew J. Murphy
Sean Stevens
George D. Yancopoulos
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Institute for Research in Biomedicine IRB
Yale University
Regeneron Pharmaceuticals Inc
Original Assignee
Institute for Research in Biomedicine IRB
Yale University
Regeneron Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Institute for Research in Biomedicine IRB, Yale University, Regeneron Pharmaceuticals Inc filed Critical Institute for Research in Biomedicine IRB
Priority to DK19176252.5T priority Critical patent/DK3556206T3/da
Priority to EP19176252.5A priority patent/EP3556206B1/en
Priority to EP21176673.8A priority patent/EP3939423B1/en
Publication of EP3308641A1 publication Critical patent/EP3308641A1/en
Application granted granted Critical
Publication of EP3308641B1 publication Critical patent/EP3308641B1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New or modified breeds of animals
    • A01K67/027New or modified breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • A01K67/0276Knock-out vertebrates
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New or modified breeds of animals
    • A01K67/027New or modified breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • A01K67/0278Knock-in vertebrates, e.g. humanised vertebrates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/0004Screening or testing of compounds for diagnosis of disorders, assessment of conditions, e.g. renal clearance, gastric emptying, testing for diabetes, allergy, rheuma, pancreas functions
    • A61K49/0008Screening agents using (non-human) animal models or transgenic animal models or chimeric hosts, e.g. Alzheimer disease animal model, transgenic model for heart failure
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/8509Vectors or expression systems specially adapted for eukaryotic hosts for animal cells for producing genetically modified animals, e.g. transgenic
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2207/00Modified animals
    • A01K2207/12Animals modified by administration of exogenous cells
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2207/00Modified animals
    • A01K2207/15Humanized animals
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • A01K2217/052Animals comprising random inserted nucleic acids (transgenic) inducing gain of function
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/072Animals genetically altered by homologous recombination maintaining or altering function, i.e. knock in
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/075Animals genetically altered by homologous recombination inducing loss of function, i.e. knock out
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/15Animals comprising multiple alterations of the genome, by transgenesis or homologous recombination, e.g. obtained by cross-breeding
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2227/00Animals characterised by species
    • A01K2227/10Mammal
    • A01K2227/105Murine
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/01Animal expressing industrially exogenous proteins
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/0331Animal model for proliferative diseases
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/524Thrombopoietin, i.e. C-MPL ligand
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/53Colony-stimulating factor [CSF]
    • C07K14/535Granulocyte CSF; Granulocyte-macrophage CSF
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5403IL-3
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5412IL-6
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/8509Vectors or expression systems specially adapted for eukaryotic hosts for animal cells for producing genetically modified animals, e.g. transgenic
    • C12N2015/8527Vectors or expression systems specially adapted for eukaryotic hosts for animal cells for producing genetically modified animals, e.g. transgenic for producing animal models, e.g. for tests or diseases
    • C12N2015/8572Animal models for proliferative diseases, e.g. comprising an oncogene

Definitions

  • MM multiple myeloma
  • BM bone marrow
  • MM is preceded by a premalignant condition called monoclonal gammopathy of undetermined significance (MGUS) that affects around 3% of persons older than 50 years ( Landgren et al., 2009, Blood 113:5412-7 ).
  • MGUS monoclonal gammopathy of undetermined significance
  • Complex heterogeneous genetic abnormalities characterize MM cells including changes in the karyotype as well as IgH translocations ( Kuehl and Bergsagel, 2002, Nat Rev Cancer. 2:175-87 ; Zhan et al., 2006, Blood 108:2020-8 ).
  • Plasma cell clones that are amplified in MGUS are thought to have genetic and phenotypic profiles similar to myelomatous plasma cells ( Chng et al., 2005, Blood 106:2156-61 ; Fonseca et al., 2002, Blood 100:1417-24 ; Kaufmann et al., 2004, Leukemia. 18:1879-82 ), While mutations in cyclin D genes have been suggested to drive development of MM, the potential contributions of other factors have not been conclusively demonstrated ( Bergsagel et al., 2005, Blood 106:296-303 ). Nonetheless, heritable genetic alterations are not the sole determinants of the behavior of MM cells. Instead, resistance towards drugs and aberrant biological responses towards cytokines are strongly influenced by interactions with the microenvironment offering an opportunity to develop novel therapeutics.
  • MM is characterized by heterogeneous cell populations strongly interacting with non-malignant stroma cells that create a supportive environment ( De Raeve and Vanderkerken, 2005, Histol Histopathol. 20:1227-50 ; Dhodapkar, 2009, Am J Hematol. 84:395-6 ).
  • the BM microenvironment for MM cells consists of a diverse extracellular matrix (ECM) and of cellular components of both hematopoietic and non-hematopoietic origin.
  • ECM extracellular matrix
  • MM pathogenesis While the BM provides a protected environment for normal hematopoiesis, the interaction of MM cells with ECM proteins and accessory cells plays a crucial role in MM pathogenesis ( De Raeve and Vanderkerken, 2005, Histol Histopathol. 20:1227-50 ; Dhodapkar, 2009, Am J Hematol. 84:395-6 ; Hideshima et al., 2007, Nat Rev Cancer. 7:585-98 ).
  • Stroma cells, myeloid cells, osteoclasts, and osteoblasts produce growth factors such as interleukin 6 (IL-6), B-cell activating factor (BAFF), fibroblast growth factor, and stroma cell-derived factor la that activate signal pathways mediating migration, survival, and growth of MM cells.
  • IL-6 interleukin 6
  • BAFF B-cell activating factor
  • fibroblast growth factor fibroblast growth factor
  • stroma cell-derived factor la stroma cell-derived factor la that activate signal pathways mediating migration, survival, and growth of MM cells.
  • IL-6 interleukin 6
  • BAFF B-cell activating factor
  • fibroblast growth factor fibroblast growth factor
  • stroma cell-derived factor la stroma cell-derived factor la that activate signal pathways mediating migration, survival, and growth of MM cells.
  • IL-6 interleukin 6
  • BAFF B-cell activating factor
  • fibroblast growth factor fibroblast growth factor
  • stroma cell-derived factor la stroma cell
  • osteoclasts and dendritic cells produce BAFF and/or a proliferation-inducing ligand (APRIL) providing anti-apoptotic signals that also increase drug resistance ( De Raeve and Vanderkerken, 2005, Histol Histopathol. 20:1227-50 ; Kukreja et al., 2006, J Exp Med. 203:1859-65 ).
  • APRIL proliferation-inducing ligand
  • mice that lack B cells, T cells, and NK cells.
  • engraftment of primary myeloma cells into these mice has been unsuccessful, but primary myeloma cells are able to engraft human fetal bone pieces upon co-transplantation into immunocompromised mice ( Yaccoby et al., 1998, Blood 92:2908-13 ).
  • MM cells are found in the human bone, but are not detected in the mouse bone or in the periphery demonstrating high residual xenorejection and a need for the human BM microenvironment ( Yaccoby et al., 1998, Blood 92:2908-13 ; Yaccoby and Epstein, 1999, Blood 94:3576-82 ).
  • NOD/Scid/ ⁇ c -/- mice allow the engraftment of several MM cell lines ( Dewan et al., 2004, Cancer Sci. 95:564-8 ; Miyakawa et al., 2004, Biochem Biophys Res Commun. 313:258-62 ).
  • the genetically modified rodents are provided that may be used to model human hematopoietic cell development, function, or disease.
  • the genetically modified rodents comprise a nucleic acid encoding human IL-6 operably linked to an IL-6 promoter and expresse at least one of human M-CSF, human IL-3, human GM-CSF, human SIRPa or human TPO.
  • the genetically modified rodent is immunodeficient.
  • the rodent is engrafted with healthy or diseased human hematopoietic cells.
  • the genetically modified rodent does not express a functional native IL-6.
  • the rodent is a mouse.
  • the IL-6 promoter to which the nucleic acid encoding human IL-6 is operably linked is the mouse IL-6 promoter, and the human IL-6 gene is operably linked to the mouse IL-6 promoter at the mouse IL-6 locus.
  • the genetically modified rodent further comprises one or more additional nucleic acids selected from a nucleic acid encoding human SIRPa under the control of a SIRPa promoter; a nucleic acid encoding human M-CSF operably linked to an M-CSF promoter, wherein the rodent expresses human M-CSF; a nucleic acid encoding human IL-3 operably linked to an IL-3 promoter, wherein the rodent expresses human IL-3; a nucleic acid encoding human GM-CSF operably linked to a GM-CSF promoter, wherein the rodent expresses human GM-CSF; and a nucleic acid encoding human TPO operably linked to a TPO promoter, wherein the rodent expresses human TPO.
  • the promoter is the human promoter for the gene. In other embodiments, the promoter is the rodent promoter for the gene. In some embodiments, the genetically modified rodent expresses the rodent's corresponding native protein. In other embodiments, the genetically modified rodent does not express the rodent's corresponding native protein.
  • the genetically modified rodent is immunodeficient for the rodent immune system.
  • the immunodeficient genetically modified rodent does not express a recombination activating gene (RAG).
  • the immunodeficient genetically modified rodent does not express the IL2 receptor gamma chain (IL2rg, or "yc").
  • the immunodeficient genetically modified rodent does not express either a RAG (e.g. RAG1, RAG2) or IL2rg.
  • the immunodeficient genetically modified rodent is engrafted with human hematopoietic cells to form a genetically modified and engrafted rodent.
  • the human hematopoietic cells are selected from human umbilical cord blood cells, human fetal liver cells, and cells of a human hematopoietic cell line.
  • the human hematopoietic cells are CD34+ progenitor cells.
  • the human hematopoietic cells are cancer cells.
  • the cancer cells are human multiple myeloma cells.
  • the genetically modified and engrafted rodent may give rise to a human cell selected from a CD34+ cell, a hematopoietic stem cell, a hematopoeitic cell, a myeloid precursor cell, a myeloid cell, a dendritic cell, a monocyte, a granulocyte, a neutrophil, a mast cell, a thymocyte, a T cell, a B cell, a plasma cell, a platelet, and a combination thereof.
  • the human cell may be present at 1 month, at 2 months, at 3 months, at 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after engraftment.
  • the genetically modified and engrafted rodent may give rise to a human hemato-lymphoid system that comprises human hematopoietic stem and progenitor cells, human myeloid progenitor cells, human myeloid cells, human dendritic cells, human monocytes, human granulocytes, human neutrophils, human mast cells, human thymocytes, human T cells, human B cells, human plasma cells, and human platelets.
  • the human hemato-lymphoid system may be present at 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after engraftment.
  • the methods comprise transplanting a population of human hematopoietic cells to a genetically modified rodent that is immunodeficient and expresses a nucleic acid encoding human IL-6 operably linked to an IL-6 promoter.
  • the animal does not express a functional native IL-6.
  • the IL-6 promoter is the rodent IL-6 promoter and the human IL-6 gene is operably linked to the rodent IL-6 promoter at the rodent IL-6 locus.
  • the rodent is a mouse.
  • the IL-6 promoter to which the nucleic acid encoding human IL-6 is operably linked is the mouse IL- 6 promoter, and the human IL-6 gene is operably linked to the mouse IL-6 promoter at the mouse IL-6 locus.
  • the transplanted population of hematopoietic cells comprises CD34+ cells. In some embodiments, the transplanted population of hematopoietic cells comprises cancer cells. In some embodiments, the transplanted population of cancer cells comprises multiple myeloma cells. In some embodiments, the transplanting comprises intrafemoral and/or intratibial injection.
  • the immunodeficient, genetically modified rodent expresses at least one additional human nucleic acid selected from the group consisting of a nucleic acid encoding human SIRPa operably linked to a SIRPa promoter; a nucleic acid encoding human M-CSF operably linked to a M-CSF promoter; a nucleic acid encoding human IL-3 operably linked to an IL-3 promoter; a nucleic acid encoding human GM-CSF operably linked to a GM-CSF promoter; and a nucleic acid encoding human TPO operably linked to a TPO promoter.
  • engrafted, genetically modified rodent expressing a nucleic acid encoding human IL-6 operably linked to an IL-6 promoter are provided, these engrafted, rodents having been prepared according to the methods described herein or as known in the art.
  • the engrafted, genetically modified rodent is an animal model of human B cell development and differentiation
  • methods that encompass the use of human hematopoietic cell-engrafted, genetically modified rodents of the subject disclosure. These methods include, for example, methods for the in vivo evaluation of the growth and differentiation of hematopoietic and immune cells, methods for the in vivo evaluation of human hematopoiesis, methods for the in vivo evaluation of cancer cells, methods for the in vivo assessment of an immune response, methods for the in vivo evaluation of vaccines and vaccination regimens, methods for the use in testing the effect of agents that modulate cancer cell growth or survival, methods for the in vivo evaluation of a treatment of cancer, and for use in testing the effect of agents that modulate hematopoietic and immune cell function.
  • methods are provided for the in vivo production and collection of human antibodies.
  • methods are provided for screening a candidate agent for the ability to treat a hematopoietic cancer.
  • the method comprises contacting a genetically modified rodent of the present disclosure that has been engrafted with human hematopoietic cancer cells with a candidate agent, and comparing the viability and/or proliferative rate of human hematopoietic cancer cells in the contacted engrafted, genetically modified rodent to the human hematopoietic cancer cells in a similarly engrafted, genetically modified rodent that is not contacted with candidate agent, wherein a decrease in the viability and/or rate of proliferation of the human hematopoietic cancer cells in the contacted engrafted, rodent indicates that the candidate agent will treat a hematopoietic cancer.
  • Genetically modified rodents are provided that may be used to model human hematopoietic cell development, function, or disease.
  • the genetically modified rodents comprise a nucleic acid encoding human IL-6 operably linked to an IL-6 promote and also express at least one of human M-CSF, human IL-3, human GM-CSF, human SIRPa or human TPO.
  • the invention also relates to methods of generating and methods of using the genetically modified rodents described herein.
  • the genetically modified rodent is a mouse.
  • the genetically modified rodent described herein is engrafted with human hematopoietic cells, including either normal or neoplastic cells, or combinations thereof.
  • the genetically modified rodent described herein is engrafted with human multiple myeloma (MM) cells.
  • the human hematopoietic cell engrafted, genetically modified rodent of the invention are useful for the in vivo evaluation of the growth and differentiation of hematopoietic and immune cells, for the in vivo evaluation of human hematopoiesis, for the in vivo evaluation of cancer cells, for the in vivo assessment of an immune response, for the in vivo evaluation of vaccines and vaccination regimens, for the use in testing the effect of agents that modulate cancer cell growth or survival, for the in vivo evaluation of a treatment of cancer, for the in vivo production and collection of immune mediators, including human antibodies, and for use in testing the effect of agents that modulate hematopoietic and immune cell function.
  • an element means one element or more than one element.
  • “About” as used herein when referring to a measurable value such as an amount, a temporal duration, and the like, is meant to encompass variations of ⁇ 20% or ⁇ 10%, more preferably ⁇ 5%, even more preferably ⁇ 1%, and still more preferably ⁇ 0.1% from the specified value, as such variations are appropriate to perform the disclosed methods.
  • abnormal when used in the context of organisms, tissues, cells or components thereof, includes those organisms, tissues, cells or components thereof that differ in at least one observable or detectable characteristic (e.g., age, treatment, time of day, etc.) from those organisms, tissues, cells or components thereof that display the "normal” (expected) respective characteristic. Characteristics which are normal or expected for one cell or tissue type, might be abnormal for a different cell or tissue type.
  • antibody includes an immunoglobulin molecule which is able to specifically bind to a specific epitope on an antigen.
  • Antibodies can be intact immunoglobulins derived from natural sources or from recombinant sources and can be immunoreactive portions of intact immunoglobulins.
  • the antibodies in the present invention may exist in a variety of forms including, for example, polyclonal antibodies, monoclonal antibodies, intracellular antibodies (“intrabodies”), Fv, Fab and F(ab)2, as well as single chain antibodies (scFv), heavy chain antibodies, such as camelid antibodies, and humanized antibodies ( Harlow et al., 1999, Using Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, NY ; Harlow et al., 1989, Antibodies: A Laboratory Manual, Cold Spring Harbor, New York ; Houston et al., 1988, Proc. Natl. Acad. Sci. USA 85:5879-5883 ; Bird et al., 1988. Science 242:423-426 ).
  • Constant expression includes a state in which a gene product is produced in a living cell under most or all physiological conditions of the cell.
  • a "coding region" of a gene includes the nucleotide residues of the coding strand of the gene and the nucleotides of the non-coding strand of the gene which are homologous with or complementary to, respectively, the coding region of an mRNA molecule which is produced by transcription of the gene.
  • a "coding region" of a mRNA molecule also includes the nucleotide residues of the mRNA molecule which are matched with an anti-codon region of a transfer RNA molecule during translation of the mRNA molecule or which encode a stop codon.
  • the coding region may thus include nucleotide residues comprising codons for amino acid residues which are not present in the mature protein encoded by the mRNA molecule (e.g., amino acid residues in a protein export signal sequence).
  • a "disease” includes a state of health of an animal wherein the animal cannot maintain homeostasis, and wherein if the disease is not ameliorated then the animal's health continues to deteriorate.
  • a "disorder" in an animal includes a state of health in which the animal is able to maintain homeostasis, but in which the animal's state of health is less favorable than it would be in the absence of the disorder. Left untreated, a disorder does not necessarily cause a further decrease in the animal's state of health.
  • a disease or disorder is "alleviated” if the severity of a symptom of the disease or disorder, the frequency with which such a symptom is experienced by a patient, or both, is reduced.
  • an “effective amount” or “therapeutically effective amount” of a compound includes that amount of compound which is sufficient to provide a beneficial effect to the subject to which the compound is administered.
  • An “effective amount” of a delivery vehicle includes that amount sufficient to effectively bind or deliver a compound.
  • Encoding includes the inherent property of specific sequences of nucleotides in a polynucleotide, such as a gene, a cDNA, or an mRNA, to serve as templates for synthesis of other polymers and macromolecules in biological processes having either a defined sequence of nucleotides (i.e., rRNA, tRNA and mRNA) or a defined sequence of amino acids and the biological properties resulting therefrom.
  • a gene encodes a protein if, for example, transcription and translation of mRNA corresponding to that gene produces the protein in a cell or other biological system.
  • Both the coding strand the nucleotide sequence of which is identical to the mRNA sequence and is usually provided in sequence listings, and the non-coding strand, used as the template for transcription of a gene or cDNA, can be referred to as encoding the protein or other product of that gene or cDNA.
  • endogenous includes any material from or produced inside an organism, cell, tissue or system.
  • exogenous includes any material introduced from or produced outside an organism, cell, tissue or system.
  • expression construct and "expression cassette” as used herein include a double-stranded recombinant DNA molecule containing a desired nucleic acid human coding sequence and containing one or more regulatory elements necessary or desirable for the expression of the operably linked coding sequence.
  • fragment includes a subsequence of a larger nucleic acid or polypeptide.
  • a “fragment” of a nucleic acid can be at least about 15 nucleotides in length; for example, at least about 50 nucleotides to about 100 nucleotides; at least about 100 to about 500 nucleotides, at least about 500 to about 1000 nucleotides, at least about 1000 nucleotides to about 1500 nucleotides; or about 1500 nucleotides to about 2500 nucleotides; or about 2500 nucleotides (and any integer value in between).
  • a "fragment" of a polypeptide can be at least about 15 nucleotides in length; for example, at least about 50 amino acids to about 100 amino acids; at least about 100 to about 500 amino acids, at least about 500 to about 1000 amino acids, at least about 1000 amino acids to about 1500 amino acids; or about 1500 amino acids to about 2500 amino acids; or about 2500 amino acids (and any integer value in between).
  • the terms "gene” and “recombinant gene” includes nucleic acid molecules comprising an open reading frame encoding a polypeptide.
  • Such natural allelic variations can typically result in 1-5% variance in the nucleotide sequence of a given gene.
  • Alternative alleles can be identified by sequencing the gene of interest in a number of different individuals. This can be readily carried out by using hybridization probes to identify the same genetic locus in a variety of individuals. Any and all such nucleotide variations and resulting amino acid polymorphisms or variations that are the result of natural allelic variation and that do not alter the functional activity are intended to be within the scope of the invention.
  • Homologous includes the subunit sequence similarity between two polymeric molecules, e.g. between two nucleic acid molecules, e.g., two DNA molecules or two RNA molecules, or between two polypeptide molecules. When a subunit position in both of the two molecules is occupied by the same monomeric subunit, e.g., if a position in each of two DNA molecules is occupied by adenine, then they are homologous at that position. The homology between two sequences is a direct function of the number of matching or homologous positions, e.g.
  • the two sequences are 50% homologous, if 90% of the positions, e.g. 9 of 10, are matched or homologous, the two sequences share 90% homology.
  • the DNA sequences 5'-ATTGCC-3' and 5'-TATGGC-3' share 50% homology.
  • human hematopoietic stem and progenitor cells include human self-renewing multipotent hematopoietic stem cells and hematopoietic progenitor cells.
  • “Inducible” expression includes a state in which a gene product is produced in a living cell in response to the presence of a signal in the cell.
  • an "instructional material” includes a publication, a recording, a diagram, or any other medium of expression which can be used to communicate the usefulness of a compound, composition, vector, or delivery system of the invention in the kit for effecting alleviation of the various diseases or disorders recited herein.
  • the instructional material can describe one or more methods of alleviating the diseases or disorders in a cell or a tissue of a mammal.
  • the instructional material of the kit of the invention can. for example, be affixed to a container which contains the identified compound, composition, vector, or delivery system of the invention or be shipped together with a container which contains the identified compound, composition, vector, or delivery system.
  • the instructional material can be shipped separately from the container with the intention that the instructional material and the compound be used cooperatively by the recipient.
  • nucleic acid includes RNA or DNA molecules having more than one nucleotide in any form including single-stranded, double-stranded, oligonucleotide or polynucleotide.
  • nucleotide sequence includes the ordering of nucleotides in an oligonucleotide or polynucleotide in a single-stranded form of nucleic acid.
  • operably linked includes a polynucleotide in functional relationship with a second polynucleotide, e.g. a single-stranded or double-stranded nucleic acid moiety comprising the two polynucleotides arranged within the nucleic acid moiety in such a manner that at least one of the two polynucleotides is able to exert a physiological effect by which it is characterized, upon the other.
  • a promoter operably linked to the coding region of a gene is able to promote transcription of the coding region.
  • the nucleic acid encoding the desired protein further comprises a promoter/regulatory sequence
  • the promoter/regulatory sequence is positioned at the 5' end of the desired protein coding sequence such that it drives expression of the desired protein in a cell.
  • the nucleic acid encoding the desired protein and its promoter/regulatory sequence comprise a "transgene.”
  • nucleotide as used herein includes a chain of nucleotides.
  • nucleic acids are polymers of nucleotides.
  • nucleic acids and polynucleotides as used herein are interchangeable.
  • nucleic acids are polynucleotides, which can be hydrolyzed into the monomeric “nucleotides.” The monomeric nucleotides can be hydrolyzed into nucleosides.
  • polynucleotides include, but are not limited to, all nucleic acid sequences which are obtained by any means available in the art, including, without limitation, recombinant means, i.e., the cloning of nucleic acid sequences from a recombinant library or a cell genome, using ordinary cloning technology and PCR, and the like, and by synthetic means.
  • recombinant means i.e., the cloning of nucleic acid sequences from a recombinant library or a cell genome, using ordinary cloning technology and PCR, and the like, and by synthetic means.
  • peptide As used herein, the terms “peptide,” “polypeptide,” and “protein” are used interchangeably, and include a compound comprised of amino acid residues covalently linked by peptide bonds.
  • a protein or peptide must contain at least two amino acids, and no limitation is placed on the maximum number of amino acids that can comprise a protein's or peptide's sequence.
  • Polypeptides include any peptide or protein comprising two or more amino acids joined to each other by peptide bonds.
  • the term includes both short chains, which also commonly are referred to in the art as peptides, oligopeptides and oligomers, for example, and to longer chains,of which there are many types.
  • Polypeptides include, for example, biologically active fragments, substantially homologous polypeptides, oligopeptides, homodimers, heterodimers, variants of polypeptides, modified polypeptides, derivatives, analogs, fusion proteins, among others.
  • the polypeptides include natural peptides, recombinant peptides, synthetic peptides, or a combination thereof.
  • peptide typically refers to short polypeptides.
  • protein typically refers to large polypeptides.
  • progeny as used herein includes a descendent or offspring and includes the differentiated or undifferentiated decedent cell derived from a parent cell. In one usage, the term progeny includes a descendent cell which is genetically identical to the parent. In another use, the term progeny includes a descendent cell which is genetically and phenotypically identical to the parent. In yet another usage, the term progeny includes a descendent cell that has differentiated from the parent cell.
  • promoter includes a DNA sequence operably linked to a nucleic acid sequence to be transcribed such as a nucleic acid sequence encoding a desired molecule.
  • a promoter is generally positioned upstream of a nucleic acid sequence to be transcribed and provides a site for specific binding by RNA polymerase and other transcription factors.
  • a promoter is generally positioned upstream of the nucleic acid sequence transcribed to produce the desired molecule, and provides a site for specific binding by RNA polymerase and other transcription factors.
  • ranges throughout this disclosure, various aspects of the invention can be presented in a range format. It should be understood that the description in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the invention. Accordingly, the description of a range should be considered to have specifically disclosed all the possible subranges as well as individual numerical values within that range. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed subranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2. 2.7, 3, 4, 5. 5.3, and 6. This applies regardless of the breadth of the range.
  • a "recombinant polypeptide” includes one which is produced upon expression of a recombinant polynucleotide.
  • regulatory element includes a nucleotide sequence which controls some aspect of the expression of nucleic acid sequences.
  • exemplary regulatory elements illustratively include an enhancer, an internal ribosome entry site (IRES), an intron; an origin of replication, a polyadenylation signal (pA), a promoter, an enhancer, a transcription termination sequence, and an upstream regulatory domain, which contribute to the replication, transcription, post-transcriptional processing of a nucleic acid sequence.
  • the term "specifically binds,” as used herein with respect to an antibody, includes an antibody which recognizes a specific antigen, but does not substantially recognize or bind other molecules in a sample.
  • an antibody that specifically binds to an antigen from one species may also bind to that antigen from one or more species. But, such cross-species reactivity does not itself alter the classification of an antibody as specific.
  • an antibody that specifically binds to an antigen may also bind to different allelic forms of the antigen. However, such cross reactivity does not itself alter the classification of an antibody as specific.
  • the terms “specific binding” or “specifically binding”, can be used in reference to the interaction of an antibody, a protein, or a peptide with a second chemical species, to mean that the interaction is dependent upon the presence of a particular structure (e.g., an antigenic determinant or epitope) on the chemical species; for example, an antibody recognizes and binds to a specific protein structure rather than to proteins generally. If an antibody is specific for epitope "A”, the presence of a molecule containing epitope A (or free, unlabeled A), in a reaction containing labeled "A” and the antibody, will reduce the amount of labeled A bound to the antibody.
  • a particular structure e.g., an antigenic determinant or epitope
  • synthetic antibody as used herein includes an antibody which is generated using recombinant DNA technology, such as, for example, an antibody expressed by a bacteriophage as described herein.
  • the term should also be construed to mean an antibody which has been generated by the synthesis of a DNA molecule encoding the antibody and which DNA molecule expresses an antibody protein, or an amino acid sequence specifying the antibody, wherein the DNA or amino acid sequence has been obtained using synthetic DNA or amino acid sequence technology which is available and well known in the art.
  • Variant includes a nucleic acid sequence or a peptide sequence that differs in sequence from a reference nucleic acid sequence or peptide sequence respectively, but retains essential biological properties of the reference molecule. Changes in the sequence of a nucleic acid variant may not alter the amino acid sequence of a peptide encoded by the reference nucleic acid, or may result in amino acid substitutions. additions, deletions, fusions and truncations. Changes in the sequence of peptide variants are typically limited or conservative, so that the sequences of the reference peptide and the variant are closely similar overall and, in many regions, identical.
  • a variant and reference peptide can differ in amino acid sequence by one or more substitutions, additions, deletions in any combination.
  • a variant of a nucleic acid or peptide can be a naturally occurring such as an allelic variant, or can be a variant that is not known to occur naturally. Non-naturally occurring variants of nucleic acids and peptides may be made by mutagenesis techniques or by direct synthesis.
  • genetically modified includes an animal, the germ cells of which comprise an exogenous human nucleic acid or human nucleic acid sequence.
  • a genetically modified animal can be a transgenic animal or a knock-in animal, so long as the animal comprises a human nucleic acid sequence.
  • transgenic animal includes an animal comprising an exogenous human nucleic acid sequence integrated into the genome of the animal.
  • knock-in includes a genetic modification that is targeted to a particular chromosomal locus of the non-human animal genome and inserts a nucleic acid of interest into that targeted locus.
  • the genetic modification replaces the genetic information encoded at the chromosomal locus in the non-human animal with a different DNA sequence.
  • a genetically modified rodent that expresses human IL-6 is provided.
  • human IL-6 hIL6 it is meant the 184 amino acid protein the sequence for which is described at, e.g, Genbank Accession Nos. NM_000600.3 and NP_000591.1.
  • Human IL-6 is a secreted protein that is produced by, for example, T cells, B cells, monocytes, macrophages, fibroblasts, keratinocytes, endothelial cells and myeloma cells.
  • IL-6 acts through a cell surface heterodimeric receptor complex comprising a binding subunit (IL-6R) and a signal transducing subunit (gp130).
  • IL-6R binding subunit
  • gp130 signal transducing subunit
  • gp130 is a common component of other receptors, such the ones for IL-11, IL-27, LIF, whereas the IL-6R is predominantly restricted to hepatocytes, monocytes, activated B cells, resting T cells and myeloma cell lines.
  • IL-6 plays a central role in hematopoiesis, in immune responses and in acute phase reactions, having been shown to be an important factor for the final maturation of B cells into antibody secreting cells (ASC), especially for the expansion of plasmablasts during the germinal center reaction in the T-dependent (TD) antibody response.
  • ASC antibody secreting cells
  • TD T-dependent
  • IL-6 is required for T cell proliferation in vitro and for generation of cytotoxic T cells (CTL) in vivo, making them more responsive to IL-2.
  • CTL cytotoxic T cells
  • the genetically modified rodent that expresses human IL-6 also expresses at least one additional human protein selected from human M-CSF, human IL-3, human GM-CSF, human TPO, and human SIRPa, or any combination thereof.
  • the rodent that expresses human IL-6 may express one, two three, four or all five of the human proteins selected from hM-CSF, hIL-3, hGM-CSF, hTPO, and hSIRPa.
  • the nucleic acid encoding the human protein is operatively linked to one or more regulatory sequences in a manner which allows for transcription of the nucleic acid into mRNA and translation of the mRNA into the human protein.
  • regulatory sequence is art-recognized and intended to include promoters, enhancers and other expression control elements (e.g., polyadenylation signals). Such regulatory sequences are known to those skilled in the art and are described in 1990, Goeddel, Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, Calif .
  • the human nucleic acid is expressed by the native regulatory elements of the human nucleic acid.
  • the human nucleic acid is expressed by the native regulatory elements of the corresponding nucleic acid of the rodent host animal.
  • the nucleic acid encoding human IL-6 is operably linked to the rodent's IL-6 promoter. In other embodiments, the nucleic acid encoding human IL-6 is operably linked to the human IL-6 promoter. As another example, in some embodiments, the nucleic acid encoding human M-CSF is operably linked to the rodent's M-CSF promoter. In other embodiments, the nucleic acid encoding human M-CSF is operably linked to the human M-CSF promoter. As a third example, in some embodiments, the nucleic acid encoding human IL-3 is operably linked to the rodent's IL-3 promoter.
  • the nucleic acid encoding human IL-3 is operably linked to the human IL-3 promoter.
  • the nucleic acid encoding human GM-CSF is operably linked to the rodent's GM-CSF promoter.
  • the nucleic acid encoding human GM-CSF is operably linked to the human GM-CSF promoter.
  • the nucleic acid encoding human TPO is operably linked to the rodent's TPO promoter.
  • the nucleic acid encoding human TPO is operably linked to the human TPO promoter.
  • the genetically modified rodents of the invention include genetically modified rodents that express at least one human nucleic acid from a promoter.
  • ubiquitously expressed promoters useful in the invention include, but are not limited to, DNA pol II promoter, PGK promoter, ubiquitin promoter, albumin promoter, globin promoter, ovalbumin promoter, SV40 early promoter, the Rous sarcoma virus (RSV) promoter, retroviral LTR and lentiviral LTR, a beta-actin promoter, a ROSA26 promoter, a heat shock protein 70 (Hsp70) promoter, an EF-1 alpha gene encoding elongation factor 1 alpha (EF1) promoter, an eukaryotic initiation factor 4A (eIF-4A1) promoter, a chloramphenicol acetyltransferase (CAT) promoter and a CMV (cytomegalovirus) promoter.
  • Promoter and enhancer expression systems useful in the invention also include inducible and/or tissue-specific expression systems.
  • tissue-specific promoters useful in the expression construct of the compositions and methods of the invention include a promoter of a gene expressed in the hematopoietic system, such as an IL-6 promoter, a M-CSF promoter, an IL-3 promoter, a GM-CSF promoter, a SIRPA promoter, a TPO promoter, an IFN- ⁇ promoter, a Wiskott-Aldrich syndrome protein (WASP) promoter, a CD45 (also called leukocyte common antigen) promoter, a Flt-1 promoter, an endoglin (CD105) promoter and an ICAM-2 (Intracellular Adhesion Molecule 2) promoter.
  • WASP Wiskott-Aldrich syndrome protein
  • promoters useful in the compositions and methods of the invention are known in the art as exemplified in Abboud et al. (2003, J. Histochem & Cytochem. 51:941-949 ), Schorpp et al. (1996, NAR 24:1787-1788 ), McBurney et al. (1994, Devel. Dynamics, 200:278-293 ) and Majumder et al. (1996, Blood 87:3203-3211 ).
  • additional regulatory elements such as an enhancer element or intron sequence, is included in various embodiments of the invention.
  • enhancers useful in the compositions and methods of the invention include, but are not limited to, a cytomegalovirus (CMV) early enhancer element and an SV40 enhancer element.
  • CMV cytomegalovirus
  • intron sequences useful in the compositions and methods of the invention include, but are not limited to, the beta globin intron or a generic intron.
  • additional regulatory elements useful in some embodiments of the invention include, but are not limited to, a transcription termination sequence and an mRNA polyadenylation (pA) sequence.
  • human nucleic acid and human amino acid encompass variants of human nucleic acid and amino acid sequences as well.
  • variant defines either an isolated naturally occurring genetic mutant of a human or a recombinantly prepared variation of a human, each of which contain one or more mutations compared with the corresponding wild-type human. For example, such mutations can be one or more amino acid substitutions, additions, and/or deletions.
  • variant also includes non-human orthologues.
  • a variant polypeptide of the present invention has at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity to a wild-type human polypeptide.
  • the percent identity between two sequences is determined using techniques as those described elsewhere herein. Mutations can be introduced using standard molecular biology techniques, such as site-directed mutagenesis and PCR-mediated mutagenesis. One of skill in the art will recognize that one or more amino acid mutations can be introduced without altering the functional properties of human proteins.
  • Conservative amino acid substitutions can be made in human proteins to produce human protein variants.
  • Conservative amino acid substitutions are art recognized substitutions of one amino acid for another amino acid having similar characteristics.
  • each amino acid may be described as having one or more of the following characteristics: electropositive, electronegative, aliphatic, aromatic, polar, hydrophobic and hydrophilic.
  • a conservative substitution is a substitution of one amino acid having a specified structural or functional characteristic for another amino acid having the same characteristic.
  • Acidic amino acids include aspartate, glutamate; basic amino acids include histidine, lysine, arginine; aliphatic amino acids include isoleucine, leucine and valine; aromatic amino acids include phenylalanine, glycine, tyrosine and tryptophan; polar amino acids include aspartate, glutamate, histidine, lysine, asparagine, glutamine, arginine, serine, threonine and tyrosine; and hydrophobic amino acids include alanine, cysteine, phenylalanine, glycine, isoleucine, leucine, methionine, proline, valine and tryptophan; and conservative substitutions include substitution among amino acids within each group. Amino acids may also be described in terms of relative size, alanine, cysteine, aspartate, glycine, asparagine, proline, threonine, serine, valine, all typically considered to be small.
  • Human variants can include synthetic amino acid analogs, amino acid derivatives and/or non-standard amino acids, illustratively including, without limitation, alpha-aminobutyric acid, citrulline, canavanine, cyanoalanine, diaminobutyric acid, diaminopimelic acid, dihydroxy-phenylalanine, djenkolic acid, homoarginine, hydroxyproline, norleucine, norvaline, 3-phosphoserine, homoserine, 5-hydroxytryptophan, 1-methylhistidine, methylhistidine, and ornithine.
  • synthetic amino acid analogs amino acid derivatives and/or non-standard amino acids
  • amino acid derivatives illustratively including, without limitation, alpha-aminobutyric acid, citrulline, canavanine, cyanoalanine, diaminobutyric acid, diaminopimelic acid, dihydroxy-phenylalanine, djenkolic acid, homoarginine, hydroxyproline
  • Human variants are encoded by nucleic acids having a high degree of identity with a nucleic acid encoding a wild-type human.
  • the complement of a nucleic acid encoding a human variant specifically hybridizes with a nucleic acid encoding a wild-type human under high stringency conditions.
  • Nucleic acids encoding a human variant can be isolated or generated recombinantly or synthetically using well-known methodology.
  • the genetically modified rodent that expresses a human nucleic acid sequence also expresses the corresponding non-human animal nucleic acid sequence.
  • the human nucleic acid sequence is randomly integrated into the genome of the rodent e.g. such that the rodent comprises the exogenous human nucleic acid sequence at a locus other than the rodent locus encoding the corresponding rodent protein.
  • the genetically modified rodent that expresses a human nucleic acid sequence does not express the corresponding rodent nucleic acid sequence.
  • the nucleic acid encoding the human protein is introduced into the rodent so as to replace genomic material encoding the corresponding rodent protein, rendering the rodent null for the corresponding rodent gene and deficient for the corresponding rodent protein.
  • the rodent is a "knock-in" for the human gene.
  • the genetically modified rodent that expresses human IL-6 also expresses rodent IL-6. In other embodiments, the genetically modified rodent that expresses human IL-6 does not express rodent IL-6.
  • the genetically modified that expresses human M-CSF also expresses rodent M-CSF. In other embodiments, the genetically modified rodent that expresses human M-CSF does not express rodent M-CSF.
  • the genetically modified rodent that expresses human IL-3 also expresses rodent IL-3. In other embodiments, the genetically modified that expresses human IL-3 does not express rodent IL-3.
  • the genetically modified rodent that expresses human GM-CSF also expresses rodent GM-CSF. In other embodiments, the genetically modified rodent that expresses human GM-CSF does not express rodent GM-CSF.
  • the genetically modified rodent that expresses human TPO also expresses rodent TPO. In other embodiments, the genetically modified rodent that expresses human TPO does not express rodent TPO.
  • the subject genetically modified rodent is immunodeficient.
  • immunodeficient it is meant that the rodent is deficient in one or more aspects of its native immune system, e.g the rodent is deficient for one or more types of functioning host immune cells, e.g. deficient for non-human B cell number and/or function, non-human T cell number and/or function, non-human NK cell number and/or function, etc.
  • the immunodeficient rodent may have severe combined immune deficiency (SCID).
  • SCID refers to a condition characterized by the absence of T cells and lack of B cell function.
  • SCID include: X-linked SCID, which is characterized by gamma chain gene mutations or loss of the IL2RG gene and the lymphocyte phenotype T(-) B(+) NK(-); and autosomal recessive SCID characterized by Jak3 gene mutations and the lymphocyte phenotype T(-) B(+) NK(-), ADA gene mutations and the lymphocyte phenotype T(-) B(-) NK(-), IL-7R alpha-chain mutations and the lymphocyte phenotype T(-) B(+) NK(+), CD3 delta or epsilon mutations and the lymphocyte phenotype T(-) B(+) NK(+), RAG1/RAG2 mutations and the lymphocyte phenotype T(-) B(-)
  • the genetically modified immunodeficient rodent has one or more deficiencies selected from an IL2 receptor gamma chain deficiency, an ADA gene mutation, an IL7R mutation, a CD3 mutation, a RAG1 and/or RAG2 mutation, an Artemis mutation, a CD45 mutation, and a Prkdc mutation.
  • the subject rodent is a mouse or a rat.
  • the rodent is selected from a mouse, a rat, and a hamster. In one embodiment, the rodent is selected from the superfamily Muroidea. In one embodiment, the genetically modified rodent is from a family selected from Calomyscidae (e.g., mouse-like hamsters), Cricetidae (e.g., hamster, New World rats and mice, voles), Muridae (true mice and rats, gerbils, spiny mice, crested rats), Nesomyidae (climbing mice, rock mice, with-tailed rats, Malagasy rats and mice), Platacanthomyidae (e.g., spiny dormice), and Spalacidae (e.g., mole rates, bamboo rats, and zokors).
  • Calomyscidae e.g., mouse-like hamsters
  • Cricetidae e.g., hamster, New World rats and mice,
  • the genetically modified rodent is selected from a true mouse or rat (family Muridae), a gerbil, a spiny mouse, and a crested rat.
  • the genetically modified mouse is from a member of the family Muridae.
  • the subject genetically modified rodent is a rat.
  • the rat is selected from a Wistar rat, an LEA strain, a Sprague Dawley strain, a Fischer strain, F344, F6, and Dark Agouti.
  • the rat strain is a mix of two or more strains selected from the group consisting of Wistar, LEA, Sprague Dawley, Fischer, F344, F6, and Dark Agouti.
  • the subject genetically modified rodent is a mouse, e.g. a mouse of a C57BL strain (e.g. C57BL/A, C57BL/An, C57BL/GrFa, C57BL/KaLwN, C57BL/6, C57BL/6J, C57BL/6ByJ, C57BL/6NJ, C57BL/10, C57BL/10ScSn, C57BL/10Cr, C57BL/Ola, etc.); a mouse of the 129 strain (e.g.
  • 129P1, 129P2, 129P3,129X1, 129S1 e.g., 129S1/SV, 129S1/SvIm),129S2, 129S4, 129S5, 129S9/SvEvH, 129S6 (129/SvEvTac), 129S7, 129S8, 129T1, 129T2; a mouse of the BALB strain; e.g., BALB/c; and the like. See, e.g., Festing et al.
  • the genetically modified mouse is a mix of an aforementioned 129 strain and an aforementioned C57BL/6 strain.
  • the mouse is a mix of aforementioned 129 strains, or a mix of aforementioned BL/6 strains.
  • the 129 strain of the mix is a 129S6 (129/SvEvTac) strain.
  • the mouse is a mix of a BALB strain and another aforementioned strain.
  • the subject genetically modified rodent is an immunodeficient mouse deficient in B cell number and/or function, and/or T cell number and/or function, and/or NK cell number and/or function (for example, due to an IL2 receptor gamma chain deficiency (i.e., ⁇ c -/- ) and/or a RAG deficiency), and having a genome that comprises a human nucleic acid, e.g. a nucleic acid encoding human IL-6, hM-CSF, hIL-3, hGM-CSF, hTPO, and/or hSIRPa, operably linked to its corresponding promoter, e.g. a M-CSF , IL-3 , GM-CSF , TPO or SIRPa promoter, respectively, wherein the rodent expresses the encoded human protein(s).
  • a human nucleic acid e.g. a nucleic acid encoding human IL-6, h
  • the subject genetically modified rodent is an immunodeficient mouse comprising a nucleic acid encoding human IL-6 operably linked to an IL-6 promoter at the mouse IL-6 locus, and a nucleic acid encoding human SIRPa operably linked to the human SIRPa promoter randomly integrated into the genome of the rodent (i.e., the mouse expresses mouse SIRPa), i.e. an immunodeficient hIL-6, hSirpa mouse, e.g.
  • the mouse further comprises a nucleic acid encoding a human M-CSF operably linked to an M-CSF promoter, a nucleic acid encoding human IL-3 operably linked to an IL-3 promoter, a nucleic acid encoding human GM-CSF operably linked to a GM-CSF promoter, and a nucleic acid encoding human TPO operably linked to a TPO promoter, i.e.
  • an immunodeficient hIL-6, hSirpa, hM-CSF, hIL-3, hGM-CSF, hTPO mouse e.g. a Rag2 -/- IL2rg -/- IL-6 h /+ M-CSF h /+ IL-3 h /+ GM-CSF h /+ TPO h /+ hSIRPa + mouse, a Rag2 - / - IL2rg -/- IL-6 h /+ M-CSF h / h IL-3 h / h GM-CSF h / h TPO h / h hSIRPa + mouse.
  • the subject genetically modified rodent is an immunodeficient mouse comprising a nucleic acid encoding human IL-6 operably linked to an IL-6 promoter and deficient for mouse IL-6, a nucleic acid encoding human SIRPa operably linked to the human SIRPa promoter randomly integrated into the genome of the non-human animal (i.e., the mouse still expresses mouse SIRPa), a nucleic acid encoding human M-CSF operably linked to an M-CSF promoter and deficient for mouse M-CSF, a nucleic acid encoding human IL-3 operably linked to an IL-3 promoter and deficient for mouse IL-3, a nucleic acid encoding human GM-CSF operably linked to a GM-CSF promoter and deficient for mouse GM-CSF, and a nucleic acid encoding human TPO operably linked to a TPO promoter and deficient for mouse TPO, i.e.
  • the subject genetically modified rodents may be generated using any convenient method for the generation of genetically modified rodents, e.g. as known in the art or as described herein.
  • a nucleic acid encoding the human protein of interest may be incorporated into a recombinant vector in a form suitable for insertion into the genome of the host cell and expression of the human protein in a non-human host cell.
  • the recombinant vector includes the one or more regulatory sequences operatively linked to the nucleic acid encoding the human protein in a manner which allows for transcription of the nucleic acid into mRNA and translation of the mRNA into the human protein, as described above.
  • the design of the vector may depend on such factors as the choice of the host cell to be transfected, the amount of human protein to be expressed, and/or how the encoding nucleic acid will integrate into the genome of the non-human host, e.g. as known in the art.
  • Any of various methods may then be used to introduce the human nucleic acid sequence into an animal cell to produce a genetically modified animal that expresses the human gene.
  • Such techniques are well-known in the art and include, but are not limited to, pronuclear microinjection of oocytes, transformation of embryonic stem cells, homologous recombination and knock-in techniques.
  • Methods for generating genetically modified animals include, but are not limited to, those described in Sundberg and Ichiki (2006, Genetically Engineered Mice Handbook, CRC Press ), Hofker and van Deursen (2002, Genetically modified Mouse Methods and Protocols, Humana Press ), Joyner (2000, Gene Targeting: A Practical Approach, Oxford University Press ), Turksen (2002, Embryonic stem cells: Methods and Protocols in Methods Mol Biol., Humana Press ), Meyer et al. (2010, Proc. Nat. Acad. Sci. USA 107:15022-15026 ), and Gibson (2004, A Primer Of Genome Science 2nd ed. Sunderland, Massachusetts: Sinauer ), U.S. Pat. No.
  • the subject genetically modified rodents can be created by introducing the nucleic acid encoding the human protein into an oocyte, e.g., by microinjection, and allowing the oocyte to develop in a female foster animal.
  • the construct comprising the human nucleic acid sequence is injected into fertilized oocytes. Fertilized oocytes can be collected from superovulated females the day after mating and injected with the expression construct. The injected oocytes are either cultured overnight or transferred directly into oviducts of 0.5-day p.c. pseudopregnant females.
  • Offspring can be evaluated for the presence of the introduced nucleic acid by DNA analysis (e.g., PCR, Southern blot, DNA sequencing, etc.) or by protein analysis (e.g., ELISA, Western blot, etc.).
  • DNA analysis e.g., PCR, Southern blot, DNA sequencing, etc.
  • protein analysis e.g., ELISA, Western blot, etc.
  • Such methods typically result in the random integration of the injected nucleic acid sequence -- in this instance, the construct comprising the nucleic acid encoding the human protein of interest - into the genome of the oocyte and hence the non-human animal, i.e. at a locus other than the locus in the host animal expressing the corresponding protein.
  • the construct comprising the nucleic acid encoding the human protein may be transfected into stem cells (ES cells or iPS cells) using well-known methods, such as electroporation, calcium-phosphate precipitation, lipofection, etc.
  • the cells can be evaluated for the presence of the introduced nucleic acid by DNA analysis (e.g., PCR, Southern blot, DNA sequencing, etc.) or by protein analysis (e.g., ELISA, Western blot, etc.). Cells determined to have incorporated the expression construct can then be introduced into preimplantation embryos.
  • Such methods are typically used in the targeted integration of the transfected nucleic acid sequence -- in this instance, the construct comprising the nucleic acid encoding the rodent human protein of interest - into the genome of the stem cells and hence the rodent. Often, such methods result in the replacement of host genomic material, e.g. genomic material encoding the corresponding host protein, with the nucleic acid encoding the human protein of interest.
  • a genetically modified founder rodent can be used to breed additional animals carrying the genetic modification.
  • Genetically modified rodents carrying a nucleic acid encoding the human protein(s) of the present disclosure can further be bred to other genetically modified rodents carrying other genetic modifications, or be bred to knockout animals, e.g., a knockout animal that does not express one or more of its genes.
  • the genetically modified immunodeficient rodents comprise a genome that includes a nucleic acid encoding a human polypeptide operably linked to a promoter, wherein the rodent expresses the encoded human polypeptide.
  • the genetically modified immunodeficient rodents comprise a genome that comprises an expression cassette that includes a nucleic acid encoding at least one human polypeptide, wherein the nucleic acid is operably linked to a promoter and a polyadenylation signal and further contains an intron, and wherein the rodent expresses the encoded human polypeptide.
  • the subject genetically modified rodent is an immunodeficient rodent.
  • Genetically modified rodents that are immunodeficient and comprise one or more human cytokines, e.g. IL-6, M-CSF, IL-3, GM-CSF, TPO, and/or SIRPa may likewise be generated using any convenient method for the generation of genetically modified rodents, e.g. as known in the art or as described herein, e.g.
  • ES embryonic stem
  • iPS induced pluripotent stem
  • Mice are then generated with the modified oocyte or ES cells using, e.g. methods described herein and known in the art, and mated to produce the immunodeficient mice comprising the desired genetic modification.
  • genetically modified rodents can be generated in a non-immunodeficient background, and crossed to an animal comprising a mutant SCID gene allele that, when homozygous, will result in immunodeficiency, and the progeny mated to create an immunodeficient animal expressing the at least one human protein of interest.
  • Various embodiments of the invention provide genetically modified rodents that include a human nucleic acid in substantially all of their cells, as well as genetically modified rodents that include a human nucleic acid in some, but not all their cells.
  • genetically modified rodents that include a human nucleic acid in some, but not all their cells.
  • one copy of the human nucleic acid will be integrated into the genome of the genetically modified rodents.
  • multiple copies, adjacent or distant to one another, of the human nucleic acid may be integrated into the genome of the genetically modified rodents.
  • the subject genetically modified rodent may be an immunodeficient rodent comprising a genome that includes a nucleic acid encoding a human polypeptide operably linked to the corresponding rodent promoter, wherein the rodent expresses the encoded human polypeptide.
  • the subject genetically modified immunodeficient rodent comprises a genome that comprises an expression cassette that includes a nucleic acid encoding at least one human polypeptide, wherein the nucleic acid is operably linked to the corresponding rodent promoter and a polyadenylation signal, and wherein the rodent expresses the encoded human polypeptide.
  • the genetically modified rodents provided in various embodiments of the present invention find many uses including, for example, for use as models of growth and differentiation of hematopoietic cells, for the in vivo evaluation of human hematopoiesis, for the in vivo evaluation of cancer cells, for in vivo study of an immune response, for in vivo evaluation of vaccines and vaccination regimens, for the use in testing the effect of agents that modulate cancer cell growth or survival, for the in vivo evaluation of a treatment of cancer, for in vivo production and collection of immune mediators, such as an antibody, and for use in testing the effect of agents that affect hematopoietic and immune cell function.
  • the subject genetically modified rodent (a "host") is engrafted with at least one human hematopoietic cell.
  • methods are provided for producing a rodent model for studies of the human hematopoietic system, comprising engrafting human hematopoietic cells into a subject genetically modified rodent (the "host”).
  • methods are provided for engrafting human hematopoietic cells into the genetically modified rodent disclosed herein.
  • the subject genetically modified rodent is engrafted with at least one human multiple myeloma cell.
  • methods are provided for producing a rodent model for cancer studies, comprising engrafting human multiple myeloma cells into a subject genetically modified rodent.
  • the invention is a method of engrafting human multiple myeloma cells into a subject genetically modified rodent.
  • the engrafted human multiple myeloma cells useful in the compositions and methods of the invention include any human multiple myeloma cell.
  • the human hematopoietic cells useful in the engraftment of the subject genetically modified rodents include any convenient human hematopoietic cell.
  • Non-limiting examples of human hematopoietic cells useful in the invention include, but are not limited to, HSC, HSPC, leukemia initiating cells (LIC), and hematopoietic cells of any lineage at any stage of differentiation, including terminally differentiated hematopoietic cells of any lineage.
  • the human hematopoietic cell is a primary cell, where "primary cells”, “primary cell lines”, and “primary cultures” are used interchangeably herein to include acutely isolated cells, or cell cultures that have been derived from a subject and allowed to grow in vitro for a limited number of passages, i.e. splittings, of the culture.
  • primary cultures are cultures that may have been passaged 0 times, 1 time, 2 times, 4 times, 5 times, 10 times, or 15 times, but not enough times go through the crisis stage.
  • the human hematopoietic cell is from a cell line, that is, the cell is from a culture that is immortalized, e.g. it has been passaged more than about 15 times.
  • the hematopoietic cells that are engrafted comprise healthy cells.
  • the hematopoietic cells that are engrafted comprise diseased hematopoietic cells, e.g. cancerous hematopoietic cells, e.g. cancerous effector B cells, i.e. multiple myeloma cells.
  • the hematopoietic cells that are engrafted comprise both healthy and diseased cells, e.g. healthy B cells and cancerous effector B cells, healthy T cells and cancerous effector B cells, etc.
  • Hematopoietic cells i.e. primary cells, cell lines generated therefrom, etc.
  • Such hematopoietic cells can be isolated from any human donor, including healthy donors, as well as donors with disease, such as cancer, including leukemia.
  • Engraftment of hematopoietic cells in the subject genetically modified rodent is characterized by the presence of human hematopoietic cells in the engrafted rodent.
  • engraftment of hematopoietic cells in the subject genetically modified rodent is characterized by the presence of differentiated human hematopoietic cells in the engrafted rodent in which hematopoietic cells are provided, as compared with appropriate control animals.
  • Hematopoietic cells including either normal and neoplastic cells, or combinations thereof, for administration to a host animal can be obtained from any tissue containing hematopoietic cells such as, but not limited to, umbilical cord blood, bone marrow, peripheral blood, cytokine or chemotherapy-mobilized peripheral blood and fetal liver.
  • Exemplary methods of isolating human hematopoietic cells, of administering human hematopoietic cells to a host animal, and of assessing engraftment of the human hematopoietic cells in the host animal are described herein and in Pearson et al. (2008, Curr. Protoc. Immunol. 81:1-15 ), Ito et al. (2002, Blood 100:3175-3182 ), Traggiai et al. (2004, Science 304:104-107 ), Ishikawa et al. (2005, Blood 106:1565-1573 ), Shultz et al. (2005, J. Immunol. 174:6477-6489 ) and Holyoake et al. (1999, Exp Hematol. 27:1418-27 ).
  • the human hematopoietic cells including either normal and neoplastic cells, or combinations thereof, are isolated from an original source material to obtain a population of cells enriched for a particular hematopoietic cell population (e.g., HSCs, HSPCs, LICs, CD34+, CD34-, lineage specific marker, cancer cell marker, etc.).
  • the isolated hematopoietic cells may or may not be a pure population.
  • hematopoietic cells useful in the compositions and methods of the invention are depleted of cells having a particular marker.
  • hematopoietic cells useful in the compositions and methods of the invention are enriched by selection for a marker.
  • hematopoietic cells useful in the compositions and methods of the invention are a population of cells in which the selected cells constitute about 1-100% of the cells, although in certain embodiments, a population of cells in which the selected cells constitute fewer than 1% of total cells can also be used.
  • hematopoietic cells useful in the compositions and methods of the invention are depleted of cells having a particular marker, such as CD34.
  • hematopoietic cells useful in the compositions and methods of the invention are enriched by selection for a marker, such as CD34.
  • hematopoietic cells useful in the compositions and methods of the invention are a population of cells in which CD34+ cells constitute about 1-100% of the cells, although in certain embodiments, a population of cells in which CD34+ cells constitute fewer than 1 % of total cells can also be used.
  • the hematopoietic cells useful in the compositions and methods of the invention are a T cell-depleted population of cells in which CD34+ cells make up about 1-3% of total cells, a lineage-depleted population of cells in which CD34+ cells make up about 50% of total cells, or a CD34+ positive selected population of cells in which CD34+ cells make up about 90% of total cells.
  • the number of hematopoietic cells administered is not considered limiting with regard to the generation of a human hematopoietic and/or immune system in a genetically modified non-human animal expressing at least one human gene.
  • the number of hematopoietic cells administered can range from about 1X10 3 to about 1X10 7 , although in various embodiments, more or fewer can also be used.
  • the number of HSPCs administered can range from about 3X10 3 to about 1X10 6 CD34+ cells when the recipient is a mouse, although in various embodiments, more or fewer can also be used.
  • the number of cells that need to be administered can be determined using only routine experimentation.
  • the genetically modified and treated mouse is engrafted with human hematopoietic cells or human hematopoietic stem cells (HPSCs) to form a genetically modified and engrafted mouse.
  • the hematopoietic cells are selected from human umbilical cord blood cells and human fetal liver cells.
  • engraftment is with about 1-2 x 10 5 human CD34+ cells.
  • administration of the hematopoietic cells may be preceded by conditioning, e.g. either sub-lethal irradiation of the recipient animal with high frequency electromagnetic radiation, generally using gamma or X-ray radiation, or treatment with a radiomimetic drug such as busulfan or nitrogen mustard.
  • conditioning is believed to reduce numbers of host hematopoietic cells, create appropriate microenvironmental factors for engraftment of human hematopoietic cells, and/or create microenvironmental niches for engraftment of human hematopoietic cells. Standard methods for conditioning are known in the art, such as described herein and in J.
  • the genetically modified mouse is treated so as to eliminate endogenous hematopoietic cells that may exist in the mouse.
  • the treatment comprises irradiating the genetically modified mouse.
  • newborn genetically modified mouse pups are irradated sublethally.
  • newborn pups are irradiated 2 x 200 cGy with a four hour interval.
  • Hematopoietic cells e.g., normal or neoplastic
  • routes such as, but not limited to, intravenous, intrahepatic, intraperitoneal, intrafemoral and/or intratibial.
  • Methods for engraftment of human hematopoietic cells, including either normal and neoplastic cells, or combinations thereof, in immunodeficient rodents are provided according to embodiments of the present invention which include providing human hematopoietic cells to the immunodeficient rodents, with or without irradiating the rodents prior to administration of the hematopoietic cells.
  • Methods for engraftment of human hematopoietic cells in immunodeficient animals are provided according to embodiments of the present invention which include providing human hematopoietic cells, including either normal and neoplastic cells, or combinations thereof, to the genetically modified rodents of the invention, with or without, administering a radiomimetic drug, such as busulfan or nitrogen mustard, to the rodents prior to administration of the hematopoietic cells.
  • a radiomimetic drug such as busulfan or nitrogen mustard
  • Engraftment of human hematopoietic cells, including either normal and neoplastic cells, or combinations thereof, in the genetically modified rodent of the invention can be assessed by any of various methods, such as, but not limited to, flow cytometric analysis of cells in the animals to which the human hematopoietic cells are administered at one or more time points following the administration of hematopoietic cells.
  • engraftment can be considered successful when the number (or percentage) of human hematopoietic cells, including either normal and neoplastic cells, or combinations thereof, present in the genetically modified rodent is greater than the number (or percentage) of human cells that were administered to the rodent at a point in time beyond the lifespan of the administered human hematopoietic cells.
  • Detection of the progeny of the administered hematopoietic cells can be achieved by detection of human DNA in the recipient animal, for example, or by detection of intact human hematopoietic cells, such as by the detection of the human cell marker, such as human CD45, human CD34, or sIL-6R for example.
  • Engraftment can be detected by flow cytometry as 0.05% or greater human CD45+ cells in the blood, spleen or bone marrow at 1-4 months after administration of the human hematopoietic cells.
  • a cytokine e.g., GM-CSF
  • GM-CSF can be used to mobilize stem cells, for example, as described in Watanabe (1997, Bone Marrow Transplantation 19:1175-1181 ).
  • the immunodeficient genetically modified and engrafted rodent gives rise to a human cell selected from a CD34+ cell, a hematopoietic stem cell, a hematopoeitic cell, a myeloid precursor cell, a myeloid cell, a dendritic cell, a monocyte, a granulocyte, a neutrophil, a mast cell, a thymocyte, a T cell, a B cell, a platelet, and a combination thereof.
  • the human cell is present at 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after engraftment.
  • the immunodeficient genetically modified and engrafted rodent gives rise to a human hemato-lymphoid system that comprises human hematopoietic stem and progenitor cells, human myeloid progenitor cells, human myeloid cells, human dendritic cells, human monocytes, human granulocytes, human neutrophils, human mast cells, human thymocytes, human T cells, human B cells, and human platelets.
  • the human hemato-lymphoid system is present at 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after engraftment.
  • the immunodeficient genetically modified and engrafted rodent gives rise to a human hemato-lymphoid system that comprises cancerous human hematopoietic cells, for example neoplastic plasma (effector B) cells.
  • cancerous human hematopoietic cells are present at 4 weeks, at 6 weeks, at 8 weeks, at 12 weeks, or at more than 12 weeks after engraftment.
  • the cancerous human hematopoietic cells are present at 2 weeks, at 4 weeks, at 6 weeks, at 8 weeks, at 12 weeks, or at more than 12 weeks after engraftment.
  • engrafted genetically modified rodents of the present disclosure are useful for studying the function of human hematopoietic cells in peripheral blood.
  • mice that are immunodeficient and comprise a nucleic acid encoding human IL-6 operably linked to an IL-6 promoter at the IL-6 mouse locus (e.g., Rag2 -/- IL2rg null IL-6 h / h mice, Rag2 -/- IL2rg null IL-6 h / h hSIRPa + mice, and Rag2 -/- IL2rg -/- IL-6 h / h M-CSF h / h IL-3 h / h GM-CSF h / h TPO h / h , hSIRPa +) support engraftment of human hematopoietic cells, e.g.
  • CD34 + progenitor cells into the peripheral blood and spleen better than immunodeficient mice that do not express human IL-6, i.e. Rag2 - / - IL2rg null mice.
  • these genetically modified mice promote the differentiation of human hematopoietic cells more efficiently than immunodeficient mice that do not express human IL-6.
  • these genetically modified mice better promote the differentiation of CD5+ B cells and CD27+ B cells.
  • CD5 is a protein found on a subset of IgM-secreting B cells called B-1 cells, and serves to mitigate activating signals from the B cell receptor so that the B-1 cells can only be activated by very strong stimuli (such as bacterial proteins) and not by normal tissue proteins.
  • CD27 is a marker for memory B cells. Additionally, these genetically modified mice support the development of better-functioning human hematopoietic cells than immunodeficient mice that do not express human IL-6. For example, B cells differentiate into IgG secreting plasma cells more rapidly in these genetically modified mice than in immunodeficient mice that do not express human IL-6. As such, engrafted genetically modified rodents of the present disclosure find use in studying hematopoietic cell development and function, and more particularly, B lymphocyte differentiation and function.
  • engrafted genetically modified rodents of the present disclosure are useful for studying hematopoietic cancers.
  • genetically modified mice that are immunodeficient and comprise a nucleic acid encoding human IL-6 operably linked to an IL-6 promoter at the mouse IL-6 locus, e.g.
  • mice do not.
  • Expression of human SIRPa by the genetically modified host further improves the rate and extent of engraftment observed.
  • engraftment of the multiple myeloma cells directly to bone of these immunodeficient, genetically modified mice disclosed herein reproduces the bone pathology typically associated with human multiple myeloma, e.g. bone destruction and resorption, e.g. as quantified by ⁇ CT scan.
  • engrafted genetically modified rodents of the present disclosure find use in screening candidate agents to identify those that will treat hematopoietic cancers.
  • treatment used herein to generally include obtaining a desired pharmacologic and/or physiologic effect.
  • the effect may be prophylactic in terms of completely or partially preventing a disease or symptom thereof and/or may be therapeutic in terms of a partial or complete cure for a disease and/or adverse effect attributable to the disease.
  • Treatment as used herein include any treatment of a disease in a mammal, and includes: (a) preventing the disease from occurring in a subject which may be predisposed to the disease but has not yet been diagnosed as having it; (b) inhibiting the disease, i.e., arresting its development; or (c) relieving the disease, i.e., causing regression of the disease.
  • Candidate agents of interest as therapeutics for hematopoietic cancers include those that may be administered before, during or after the onset of cancer.
  • the treatment of ongoing disease, where the treatment stabilizes or reduces the undesirable clinical symptoms of the patient, is of particular interest.
  • the terms "individual,” “subject,” “host,” and “patient,” are used interchangeably herein and include any mammalian subject for whom diagnosis, treatment, or therapy is desired, particularly humans.
  • engrafted genetically modified rodents of the present disclosure are useful for studying human pathogens, i.e. pathogens that infect humans; the response of the human immune system to infection by human pathogens; and the effectiveness of agents in protecting against and/or treating infection by human pathogens.
  • the pathogen may be a virus, a fungus, a bacterium, etc.
  • Non-limiting examples of viral pathogens include human or porcine or avian influenza virus.
  • Non-limiting examples of bacterial pathogens include mycobacterium, e.g. Mycobacterium tuberculosis (M. tuberculosis ), and enterobacterium, e.g. Salmonella typhi (S. typhi).
  • Examples of methods for infecting mice with S. typhi and for assessing infection may be found in, for example, US Published Application No. 2011/0200982 .
  • Examples of methods for infecting mice with M. tuberculosis and for assessing infection may be found in, for example, US Published Application No. 2011/0200982 .
  • Other examples of human pathogens that do not infect wild-type mice, or that infect wild-type mice but the infected mice do not model an immune response that a human mounts in response to the pathogen, will be well-known to the ordinarily skilled artisan.
  • Such mouse models of pathogen infection are useful in research, e.g. to better understand the progression of human infection.
  • Such mouse models of infection are also useful in drug discovery, e.g. to identify candidate agents that protect against or treat infection.
  • Engrafted genetically modified mice of the present disclosure also provide a useful system for screening candidate agents for desired activities in vivo, for example, to identify agents that are able to modulate ( i . e ., promote or suppress) hematopoietic cell development and/or activity, e.g. the activity of B cells, T cells, NK cells, macrophages, neutrophils, eosinophils, basophils, etc., e.g. in a healthy or a diseased state, e.g.
  • cancerous cells during pathogen infection, for example to identify novel therapeutics and/or develop a better understanding of the molecular basis of the development and function of the immune system; for agents that are toxic to hematopoietic cells, e.g. B cells, T cells, NK cells, macrophages, neutrophils, eosinophils, basophils, etc ., and progenitors thereof; and for agents that prevent against, mitigate, or reverse the toxic effects of toxic agents on hematopoietic cells, e.g. B cells, T cells, NK cells, macrophages, neutrophils, eosinophils, basophils, etc., and progenitors thereof; etc.
  • agents that are toxic to hematopoietic cells e.g. B cells, T cells, NK cells, macrophages, neutrophils, eosinophils, basophils, etc., and progenitors thereof; and for agents that prevent against, mitigate, or reverse the toxic effects of toxic agents
  • engrafted genetically modified rodents of the present disclosure provide a useful system for predicting the responsiveness of an individual to a disease therapy, e.g. by providing an in vivo platform for screening the responsiveness of an individual's immune system to an agent, e.g. a therapeutic agent, to predict the responsiveness of an individual to that agent.
  • an agent e.g. a therapeutic agent
  • a human hematopoietic cell-engrafted genetically modified mouse of the present disclosure e.g. an engrafted Rag2 - / - IL2rg - / - IL-6 h / h hSIRPa + mouse, an engrafted Rag2 - / - IL2rg - / - IL-6 h / h M-CSF h / h IL-3 h / h GM-CSF h / h TPO h / h , hSIRPa + mouse, etc.
  • an engrafted Rag2 - / - IL2rg - / - IL-6 h / hSIRPa + mouse an engrafted Rag2 - / - IL2rg - / - IL-6 h / h M-CSF h / h IL-3 h / h GM-CSF h
  • output parameters may be reflective of the viability of the cells, e.g. the total number of hematopoietic cells or the number of cells of a particular hematopoietic cell type, or of the apoptotic state of the cells, e.g. the amount of DNA fragmentation, the amount of cell blebbing, the amount of phosphatidylserine on the cell surface, and the like by methods that are well known in the art.
  • the output parameters may be reflective of the differentiation capacity of the cells, e.g. the proportions of differentiated cells and differentiated cell types.
  • the output parameters may be reflective of the function of the cells, e.g. the cytokines and chemokines produced by the cells, the antibodies (e.g. amount or type) produced by the cells, the ability of the cells to home to and extravasate to a site of challenge, the ability of the cells to modulate, i.e. promote or suppress, the activity of other cells in vitro or in vivo, etc.
  • Other output parameters may be reflective of the extent of damage induced by diseased hematopoietic cells, e.g. bone destruction and resorption induced by multiple myeloid cells.
  • Yet other parameters may be reflective of the effect of the agent on infection, e.g. pathogen infection in the animal, e.g. the titer of pathogen in the mouse, the presence of granuloma in the mouse, etc ., as relevant to the studies being performed.
  • Parameters are quantifiable components of cells, particularly components that can be accurately measured, desirably in a high throughput system.
  • a parameter can be any cell component or cell product including cell surface determinant, receptor, protein or conformational or posttranslational modification thereof, lipid, carbohydrate, organic or inorganic molecule, nucleic acid, e.g. mRNA, DNA, etc. or a portion derived from such a cell component or combinations thereof. While most parameters will provide a quantitative readout, in some instances a semi-quantitative or qualitative result will be acceptable. Readouts may include a single determined value, or may include mean, median value or the variance, etc.
  • Characteristically a range of parameter readout values will be obtained for each parameter from a multiplicity of the same assays. Variability is expected and a range of values for each of the set of test parameters will be obtained using standard statistical methods with a common statistical method used to provide single values.
  • Candidate agents of interest for screening include known and unknown compounds that encompass numerous chemical classes, primarily organic molecules, which may include organometallic molecules, inorganic molecules, genetic sequences, vaccines, antibiotics or other agents suspected of having antibiotic properties, peptides, polypeptides, antibodies, agents that have been approved pharmaceutical for use in a human, etc.
  • An important aspect of the invention is to evaluate candidate drugs, including toxicity testing; and the like.
  • Candidate agents include organic molecules comprising functional groups necessary for structural interactions, particularly hydrogen bonding, and typically include at least an amine, carbonyl, hydroxyl or carboxyl group, frequently at least two of the functional chemical groups.
  • the candidate agents often comprise cyclical carbon or heterocyclic structures and/or aromatic or polyaromatic structures substituted with one or more of the above functional groups.
  • Candidate agents are also found among biomolecules, including peptides, polynucleotides, saccharides, fatty acids, steroids, purines, pyrimidines, derivatives, structural analogs or combinations thereof. Included are pharmacologically active drugs, genetically active molecules, etc.
  • Compounds of interest include chemotherapeutic agents, hormones or hormone antagonists, etc.
  • Exemplary of pharmaceutical agents suitable for this invention are those described in, " The Pharmacological Basis of Therapeutics,” Goodman and Gilman, McGraw-Hill, New York, N.Y., (1996), Ninth editi on. Also included are toxins, and biological and chemical warfare agents, for example see Somani, S. M. (Ed.), “Chemical Warfare Agents,” Academic Press, New York, 1992 ).
  • Candidate agents of interest for screening also include nucleic acids, for example, nucleic acids that encode siRNA, shRNA, antisense molecules, or miRNA, or nucleic acids that encode polypeptides.
  • Many vectors useful for transferring nucleic acids into target cells are available.
  • the vectors may be maintained episomally, e . g . as plasmids, minicircle DNAs, virus-derived vectors such cytomegalovirus, adenovirus, etc ., or they may be integrated into the target cell genome, through homologous recombination or random integration, e.g. retrovirus derived vectors such as MMLV, HIV-1, ALV, etc.
  • Vectors may be provided directly to the subject cells. In other words, the pluripotent cells are contacted with vectors comprising the nucleic acid of interest such that the vectors are taken up by the cells.
  • nucleic acid vectors such as electroporation, calcium chloride transfection, and lipofection
  • the nucleic acid of interest may be provided to the cells via a virus.
  • the cells are contacted with viral particles comprising the nucleic acid of interest.
  • Retroviruses for example, lentiviruses, are particularly suitable to the method of the invention. Commonly used retroviral vectors are "defective", i.e. unable to produce viral proteins required for productive infection. Rather, replication of the vector requires growth in a packaging cell line.
  • the retroviral nucleic acids comprising the nucleic acid are packaged into viral capsids by a packaging cell line.
  • Different packaging cell lines provide a different envelope protein to be incorporated into the capsid, this envelope protein determining the specificity of the viral particle for the cells.
  • Envelope proteins are of at least three types, ecotropic, amphotropic and xenotropic.
  • Retroviruses packaged with ecotropic envelope protein, e.g. MMLV are capable of infecting most murine and rat cell types, and are generated by using ecotropic packaging cell lines such as BOSC23 ( Pear et al. (1993) P.N.A.S. 90:8392-8396 ).
  • Retroviruses bearing amphotropic envelope protein are capable of infecting most mammalian cell types, including human, dog and mouse, and are generated by using amphotropic packaging cell lines such as PA12 ( Miller et al. (1985) Mol. Cell. Biol. 5:431-437 ); PA317 ( Miller et al. (1986) Mol. Cell. Biol. 6:2895-2902 ); GRIP ( Danos et al. (1988) PNAS 85:6460-6464 ). Retroviruses packaged with xenotropic envelope protein, e.g. AKR env, are capable of infecting most mammalian cell types, except murine cells.
  • the appropriate packaging cell line may be used to ensure that the cells of interest-in some instance, the engrafted cells, in some instance, the cells of the host, i.e. the genetically modified animal--are targeted by the packaged viral particles.
  • Vectors used for providing nucleic acid of interest to the subject cells will typically comprise suitable promoters for driving the expression, that is, transcriptional activation, of the nucleic acid of interest.
  • suitable promoters for driving the expression that is, transcriptional activation, of the nucleic acid of interest.
  • This may include ubiquitously acting promoters, for example, the CMV- ⁇ -actin promoter, or inducible promoters, such as promoters that are active in particular cell populations or that respond to the presence of drugs such as tetracycline.
  • transcriptional activation it is intended that transcription will be increased above basal levels in the target cell by at least about 10 fold, by at least about 100 fold, more usually by at least about 1000 fold.
  • vectors used for providing reprogramming factors to the subject cells may include genes that must later be removed, e.g. using a recombinase system such as Cre/Lox, or the cells that express them destroyed, e.g. by including genes that allow selective toxicity such as herpes
  • Candidate agents of interest for screening also include polypeptides. Such polypeptides may optionally be fused to a polypeptide domain that increases solubility of the product.
  • the domain may be linked to the polypeptide through a defined protease cleavage site, e.g. a TEV sequence, which is cleaved by TEV protease.
  • the linker may also include one or more flexible sequences, e.g. from 1 to 10 glycine residues.
  • the cleavage of the fusion protein is performed in a buffer that maintains solubility of the product, e.g.
  • Domains of interest include endosomolytic domains, e.g. influenza HA domain; and other polypeptides that aid in production, e.g. IF2 domain, GST domain, GRPE domain, and the like. Additionally or alternatively, such polypeptides may be formulated for improved stability.
  • the peptides may be PEGylated. where the polyethyleneoxy group provides for enhanced lifetime in the blood stream.
  • the polypeptide may be fused to another polypeptide to provide for added functionality, e . g . to increase the in vivo stability.
  • fusion partners are a stable plasma protein, which may, for example, extend the in vivo plasma half-life of the polypeptide when present as a fusion, in particular wherein such a stable plasma protein is an immunoglobulin constant domain.
  • the stable plasma protein is normally found in a multimeric form, e.g., immunoglobulins or lipoproteins, in which the same or different polypeptide chains are normally disulfide and/or noncovalently bound to form an assembled multichain polypeptide
  • the fusions herein containing the polypeptide also will be produced and employed as a multimer having substantially the same structure as the stable plasma protein precursor.
  • These multimers will be homogeneous with respect to the polypeptide agent they comprise, or they may contain more than one polypeptide agent.
  • the candidate polypeptide agent may be produced from eukaryotic cells, or may be produced by prokaryotic cells. It may be further processed by unfolding, e.g. heat denaturation, DTT reduction, etc. and may be further refolded, using methods known in the art. Modifications of interest that do not alter primary sequence include chemical derivatization of polypeptides, e . g ., acylation, acetylation, carboxylation, amidation, etc. Also included are modifications of glycosylation, e . g . those made by modifying the glycosylation patterns of a polypeptide during its synthesis and processing or in further processing steps; e . g .
  • polypeptides by exposing the polypeptide to enzymes which affect glycosylation, such as mammalian glycosylating or deglycosylating enzymes.
  • enzymes which affect glycosylation such as mammalian glycosylating or deglycosylating enzymes.
  • sequences that have phosphorylated amino acid residues e . g . phosphotyrosine, phosphoserine, or phosphothreonine.
  • the polypeptides may have been modified using ordinary molecular biological techniques and synthetic chemistry so as to improve their resistance to proteolytic degradation or to optimize solubility properties or to render them more suitable as a therapeutic agent.
  • Analogs of such polypeptides include those containing residues other than naturally occurring L-amino acids, e.g. D-amino acids or non-naturally occurring synthetic amino acids. D-amino acids may be substituted for some or all of the amino acid residues.
  • the candidate polypeptide agent may be prepared by in vitro synthesis, using conventional methods as known in the art.
  • Various commercial synthetic apparatuses are available, for example, automated synthesizers by Applied Biosystems, Inc., Beckman, etc. By using synthesizers, naturally occurring amino acids may be substituted with unnatural amino acids. The particular sequence and the manner of preparation will be determined by convenience, economics, purity required, and the like.
  • the candidate polypeptide agent may be isolated and purified in accordance with conventional methods of recombinant synthesis.
  • a lysate may be prepared of the expression host and the lysate purified using HPLC, exclusion chromatography, gel electrophoresis, affinity chromatography, or other purification technique.
  • compositions which are used will comprise at least 20% by weight of the desired product, more usually at least about 75% by weight, preferably at least about 95% by weight, and for therapeutic purposes, usually at least about 99.5% by weight, in relation to contaminants related to the method of preparation of the product and its purification.
  • percentages will be based upon total protein.
  • the candidate polypeptide agents to be screened are antibodies.
  • antibody or “antibody moiety” is intended to include any polypeptide chain-containing molecular structure with a specific shape that fits to and recognizes an epitope, where one or more non-covalent binding interactions stabilize the complex between the molecular structure and the epitope.
  • the specific or selective fit of a given structure and its specific epitope is sometimes referred to as a "lock and key” fit.
  • the archetypal antibody molecule is the immunoglobulin, and all types of immunoglobulins, IgG, IgM, IgA, IgE, IgD, etc., from all sources, e.g.
  • Antibodies utilized in the present invention may be either polyclonal antibodies or monoclonal antibodies. Antibodies are typically provided in the media in which the cells are cultured. Antibody production and screen is discussed in greater detail below.
  • Candidate agents may be obtained from a wide variety of sources including libraries of synthetic or natural compounds. For example, numerous means are available for random and directed synthesis of a wide variety of organic compounds, including biomolecules, including expression of randomized oligonucleotides and oligopeptides. Alternatively, libraries of natural compounds in the form of bacterial, fungal, plant and animal extracts are available or readily produced. Additionally, natural or synthetically produced libraries and compounds are readily modified through conventional chemical, physical and biochemical means, and may be used to produce combinatorial libraries. Known pharmacological agents may be subjected to directed or random chemical modifications, such as acylation, alkylation, esterification, amidification, etc. to produce structural analogs.
  • Candidate agents are screened tor biological activity by administering the agent to at least one and usually a plurality of samples, sometimes in conjunction with samples lacking the agent.
  • the change in parameters in response to the agent is measured, and the result evaluated by comparison to reference cultures, e.g . in the presence and absence of the agent, obtained with other agents, etc.
  • the screen is typically performed in the presence of the toxic agent, where the toxic agent is added at the time most appropriate to the results to be determined.
  • the candidate agent may be added before the toxic agent, simultaneously with the candidate agent, or subsequent to treatment with the candidate agent.
  • the candidate agent may be added subsequent to treatment with the candidate agent.
  • the “sample” is a genetically modified non-human animal that has been engrafted with cells, e.g. the candidate agent is provided to an immunodeficient animal, e.g. mouse, comprising a nucleic acid encoding human IL-6 operably linked to an IL-6 promoter that has been engrafted with human hematopoietic cells.
  • the sample is the human hematopoietic cells to be engrafted, i.e. the candidate agent is provided to cells prior to engraftment into the immunodeficient genetically modified animal.
  • the agent may be administered by any of a number of well-known methods in the art for the administration of peptides, small molecules and nucleic acids to mice.
  • the agent may be administered orally, mucosally, topically, intrdermally, or by injection, e.g . intraperitoneal, subcutaneous, intramuscular, intravenous, or intracranial injection, and the like.
  • the agent may be administered in a buffer, or it may be incorporated into any of a variety of formulations, e.g. by combination with appropriate pharmaceutically acceptable vehicle.
  • “Pharmaceutically acceptable vehicles” may be vehicles approved by a regulatory agency of the Federal or a state government or listed in the U.S.
  • lipids e.g. liposomes, e.g. liposome dendrimers
  • liquids such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like, saline; gum acacia, gelatin, starch paste, talc, keratin, colloidal silica, urea, and the like.
  • compositions may be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants, gels, microspheres, and aerosols.
  • the agent may be systemic after administration or may be localized by the use of regional administration, intramural administration, or use of an implant that acts to retain the active dose at the site of implantation.
  • the active agent may be formulated for immediate activity or it may be formulated for sustained release. For some conditions, particularly central nervous system conditions, it may be necessary to formulate agents to cross the blood-brain barrier (BBB).
  • BBB blood-brain barrier
  • BBB blood-brain barrier
  • osmotic means such as mannitol or leukotrienes
  • vasoactive substances such as bradykinin.
  • a BBB disrupting agent can be co-administered with the agent when the compositions are administered by intravascular injection.
  • Other strategies to go through the BBB may entail the use of endogenous transport systems, including Caveolin-1 mediated transcytosis, carrier-mediated transporters such as glucose and amino acid carriers, receptor-mediated transcytosis for insulin or transferrin, and active efflux transporters such as p-glycoprotein.
  • Active transport moieties may also be conjugated to the therapeutic compounds for use in the invention to facilitate transport across the endothelial wall of the blood vessel.
  • drug delivery of agents behind the BBB may be by local delivery, for example by intrathecal delivery, e.g. through an Ommaya reservoir (see e.g. US Patent Nos. 5,222,982 and 5385582 ); by bolus injection, e.g. by a syringe, e.g. intravitreally or intracranially; by continuous infusion, e.g. by cannulation, e.g. with convection (see e.g. US Application No. 20070254842 ); or by implanting a device upon which the agent has been reversably affixed (see e.g. US Application Nos. 20080081064 and 20090196903 ).
  • the agents are conveniently added in solution, or readily soluble form, to the medium of cells in culture.
  • the agents may be added in a flow-through system, as a stream, intermittent or continuous, or alternatively, adding a bolus of the compound, singly or incrementally, to an otherwise static solution.
  • a flow-through system two fluids are used, where one is a physiologically neutral solution, and the other is the same solution with the test compound added. The first fluid is passed over the cells, followed by the second.
  • a bolus of the test compound is added to the volume of medium surrounding the cells. The overall concentrations of the components of the culture medium should not change significantly with the addition of the bolus, or between the two solutions in a flow through method.
  • a plurality of assays may be run in parallel with different agent concentrations to obtain a differential response to the various concentrations.
  • determining the effective concentration of an agent typically uses a range of concentrations resulting from 1:10, or other log scale, dilutions.
  • the concentrations may be further refined with a second series of dilutions, if necessary.
  • one of these concentrations serves as a negative control, i.e. at zero concentration or below the level of detection of the agent or at or below the concentration of agent that does not give a detectable change in the phenotype.
  • An analysis of the response of cells in the engrafted genetically modified animal to the candidate agent may be performed at any time following treatment with the agent.
  • the cells may be analyzed 1, 2, or 3 days, sometimes 4, 5, or 6 days, sometimes 8, 9, or 10 days, sometimes 14 days, sometimes 21 days, sometimes 28 days, sometimes 1 month or more after contact with the candidate agent, e.g. 2 months, 4 months, 6 months or more.
  • the analysis comprises analysis at multiple time points. The selection of the time point(s) for analysis will be based upon the type of analysis to be performed, as will be readily understood by the ordinarily skilled artisan.
  • the analysis may comprise measuring any of the parameters described herein or known in the art for measuring cell viability, cell proliferation, cell identity, cell morphology, and cell function, particularly as they may pertain to cells of the immune cells.
  • flow cytometry may be used to determine the total number of hematopoietic cells or the number of cells of a particular hematopoietic cell type.
  • Histochemistry or immunohistochemistry may be performed to determine the apoptotic state of the cells, e.g. terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) to measure DNA fragmentation, or immunohistochemistry to detect Annexin V binding to phosphatidylserine on the cell surface.
  • TUNEL terminal deoxynucleotidyl transferase dUTP nick end labeling
  • Flow cytometry may also be employed to assess the proportions of differentiated cells and differentiated cell types, e.g. to determine the ability of hematopoietic cells to differentiate in the presence of agent.
  • ELISAs, Westerns, and Northern blots may be performed to determine the levels of cytokines, chemokines, immunoglobulins, etc. expressed in the engrafted genetically modified mice, e.g. to assess the function of the engrafted cells, to assess the survival of cancerous plasma cells, etc.
  • ⁇ CT scans may be performed to determine the extent of damage induced by diseased hematopoietic cells, e.g. bone destruction and resorption induced by multiple myeloid cells.
  • In vivo assays to test the function of immune cells may also be performed. See, e.g. Current Protocols in Immunology (Richard Coico, ed. John Wiley & Sons, Inc. 2012 ) and Immunology Methods Manual (I. Lefkovits ed., Academic Press 1997 ),.
  • a method for determining the effect of an agent on multiple myeloma comprising administering the agent to a humanized IL-6 mouse, e.g. a Rag2 - / - IL2rg - / - IL-6 h / h mouse, that has been engrafted with human multiple myeloma cells; measuring a parameter of the viability and/or proliferative ability of the multiple myeloma cells over time in the presence of the agent; and comparing that measurement to the measurement from an engrafted humanized IL-6 mouse not exposed to the agent.
  • a humanized IL-6 mouse e.g. a Rag2 - / - IL2rg - / - IL-6 h / h mouse
  • the agent is determined to be anti-cancerous if it reduces the proliferation of and/or reduces the number of multiple myeloma cells in blood or a tissue of the mouse by at least 20%, 30%, 40% or more, in some instances 50%, 60%, 70% or more, e.g. 80%, 90% or 100%, i.e., to undetectable amounts, following a single administration or two or more administrations of the agent over a selected period of time.
  • the administration of the drug or combination of drugs is at least a week, 10 days, two week, three weeks, or four weeks after engraftment of the multiple myeloma cells.
  • mice Other examples of uses for the subject mice are provided elsewhere herein. Additional applications of the genetically modified and engrafted mice described in this disclosure will be apparent to those skilled in the art upon reading this disclosure.
  • compositions and methods useful for the production of human monoclonal antibodies from an engrafted immunodeficient rodent comprise contacting an immunodeficient rodent with a human hematopoietic cell to generate an immune system-transplanted rodent (engrafted rodent), subsequently contacting the engrafted rodent with an antigen, collecting from the engrafted rodent a human cell producing a human antibody against the antigen, and isolating the antibody from the antibody producing cell.
  • the invention comprises a method that includes establishing an antibody producing cell (e.g., a human B-cell) by a transformation method (e.g. EBV) or a cell fusion method (e.g. hybridoma).
  • an antibody producing cell e.g., a human B-cell
  • a transformation method e.g. EBV
  • a cell fusion method e.g. hybridoma
  • the antibody producing cell is capable of being maintained under suitable cell culture conditions for at least about 50 passages.
  • the engrafted animal is a mouse or a rat.
  • the human hematopoietic cell is CD34+ cell obtained from a human fetal liver, bone marrow, cord blood, peripheral blood, or spleen sample.
  • the antigen at least one of: a peptide, a polypeptide, an MHC/peptide complex, DNA, a live virus, a dead virus or portion thereof, a live bacteria, a dead bacteria or portion thereof, or a cancer cell or portion thereof.
  • the engrafted animal has been contacted with the antigen 1-5 months after the animal has been contacted with the human hematopoietic cell. In some embodiments, the engrafted animal is contacted only one time with the antigen, while in other embodiments, the engrafted animal is contacted two, three, four, five, six, seven, eight, or more times with the antigen.
  • human antibody producing cell collected from the engrafted animal is a B cell.
  • the human antibody producing cell collected from the animal expresses on its surface at least one of: CD19, CD20, CD22, and CD27.
  • the human antibody-producing cell of the invention can be recovered by removal of any suitable cellular components of the immune system from the animal.
  • the antibody-producing cell is removed from the engrafted animal by removal of at least one of the spleen, the lymph nodes, the peripheral blood, the bone marrow or portions thereof.
  • the method of the invention employs a conventional hybridoma technology using a suitable fusion partner.
  • the fusion partner at least one cell selected from the group consisting of: MOPC21, P3X63AG8, SP2/0, NS-1, P3.X63AG8.653, F0, S194/5.XXO.BU-1, FOX-NY, SP2/0-Ag14, MEG-01, HEL, UT-7, M07e, MEG-A2, and DAMI, and cell lines derived from these cells.
  • Isolation of the antibody from the antibody producing cell, the media in which the antibody producing cell is culture, and/or the ascites of the engrafted animal can be performed according to the methods known in the art, such as, by way of example, chromatography and dialysis.
  • the antibody can be isolated using one or more of immunoaffinity purification, ammonium sulphate precipitation, protein A/G purification, ion exchange chromatography and gel filtration.
  • immunoaffinity purification ammonium sulphate precipitation
  • protein A/G purification protein A/G purification
  • ion exchange chromatography ion exchange chromatography
  • the antigen may be administered to the engrafted rodent by any suitable means known in art.
  • the antigen can be administered to the engrafted rodent by at least one of intrasplenically, intravenously, intraperitoneally, intradermally, intramuscularly, and subcutaneously.
  • the antigen is administered alone and in other embodiments, the antigen is administered in combination with appropriate immunomodulating agent or adjuvant.
  • adjuvants useful in the methods of the invention include, but are not limited to, Complete Freund's Adjuvant (CFA), Incomplete Freund's Adjuvant (IFA), and Alum (Al 3 (OH) 4 ).
  • reagents and kits thereof for practicing one or more of the above-described methods.
  • the subject reagents and kits thereof may vary greatly.
  • the reagents or kits may comprise one or more reagents for use in the generation and/or maintenance of the subject genetically modified rodents.
  • the kit may comprise an immunodeficient mouse comprising a nucleic acid encoding human IL-6 operably linked to an IL-6 promoter and a nucleic acid encoding human SIRPa operably linked to a SIRPa promoter; or a mouse comprising a nucleic acid encoding human IL-6 operably linked to an IL-6 promoter and further comprising a nucleic acid encoding human M-CSF operably linked to an M-CSF promoter; a nucleic acid encoding human IL-3 operably linked to an IL-3 promoter; a nucleic acid encoding human GM-CSF operably linked to a GM-CSF promoter; a nucleic acid encoding human TPO operably linked to a TPO promoter; and/or a nucleic acid encoding human SIRPa operably linked to a SIRPa promoter.
  • the kit may comprise reagents for breeding such mice, e.g. primers for genotyping for the human IL-6 gene, for the human M-CSF gene, for the human IL-3 gene, for the human GM-CSF gene, for the human SIRPa gene, and/or for the human TPO gene, PCR buffer, MgCl 2 solution, etc.
  • the reagents or kits may comprise one or more reagents for use in engrafting the subject genetically modified rodents, for example human hematopoietic cells, an enriched population of human hematopoietic progenitor cells, a hematopoietic cell line, a neoplastic hematopoietic cell line, etc. for transplantation into the subject genetically modified rodents, or reagents for preparing a population of hematopoietic cells, an enriched population of hematopoietic cells from a human, a hematopoietic cell line, a neoplastic hematopoietic cell line, etc. for transplantation into a subject genetically modified rodents.
  • reagents for use in engrafting the subject genetically modified rodents for example human hematopoietic cells, an enriched population of human hematopoietic progenitor cells, a hematopoietic cell line
  • the reagents or kits may include reagents for determining the viability and/or function of hematopoietic cells, e.g. in the presence/absence of a candidate agent, e.g. one or more antibodies that are specific for markers expressed by different types of hematopoietic cells, or reagents for detecting particular cytokines, chemokine, etc..
  • a candidate agent e.g. one or more antibodies that are specific for markers expressed by different types of hematopoietic cells
  • Other reagents may include culture media, culture supplements, matrix compositions, and the like.
  • the subject kits may further include instructions for practicing the subject methods. These instructions may be present in the subject kits in a variety of forms, one or more of which may be present in the kit.
  • One form in which these instructions may be present is as printed information on a suitable medium or substrate, e.g., a piece or pieces of paper on which the information is printed, in the packaging of the kit, in a package insert, etc.
  • Yet another means would be a computer readable medium, e.g., diskette, CD, etc., on which the information has been recorded.
  • Yet another means that may be present is a website address which may be used via the internet to access the information at a removed site. Any convenient means may be present in the kits.
  • Example 1 Genetic humanization of cytokine genes enables engraftment of mice with human multiple myeloma cells.
  • the data described herein demonstrate that the genetically modified non-human animals described herein represent a novel in vivo animal model for multiple myeloma.
  • mice Humanized IL-6 knock-in mice were generated by replacing 6.8 kb of the murine IL-6 gene locus with a 4.8-kb human IL-6 gene sequence containing exons 1 through 5 including 3' untranslated region of the human IL-6 gene.
  • a targeting construct for replacing the mouse with the human IL-6 gene in a single targeting step was constructed using VELOCIGENE® genetic engineering technology (see, Valenzuela et al. (2003) High-throughput engineering of the mouse genome coupled with high-resolution expression analysis, Nature Biotech, 21(6):652-659 ).
  • Mouse and human IL-6 DNA were obtained from bacterial artificial chromosome (BAC) RPCI-23 clone 368C3, and from BAC CTD clone 2369M23, respectively.
  • BAC bacterial artificial chromosome
  • a NotI linearized targeting construct generated by gap repair cloning containing mouse IL-6 upstream and downstream homology arms flanking a 4.8 kb human IL-6 sequence extending from ATG in exon 1 to exon 5 with 16 nucleotides of 3' downstream sequence (genomic coordinates: NCBIh37.1: ch7:22,766,882 to 22,771,637) and a floxed neo selection cassette, was electroporated into Rag2 +/- IL2rg Y/- ES cells.
  • the parental ES cell line in which the RAG2 gene and IL2rg gene knockout was made was a commercially available V17 ES cell (BALB/cxl29 heterozygote). Correctly targeted ES cells may be electroporated with a transient Cre-expressing vector to remove the drug selection cassette.
  • LONA loss-of-native-allele
  • the qPCR assays comprised the following primer-probe sets (written 5' to 3'): upstream forward primer, TTGCCGGTTT TCCCTTTTCT C (SEQ ID NO:1); upstream reverse primer, AGGGAAGGCC GTGGTTGTC (SEQ ID NO:2); upstream probe, FAM-CCAGCATCAG TCCCAAGAAG GCAACT-BHQ (SEQ ID NO:3); downstream forward primer, TCAGAGTGTG GGCGAACAAA G (SEQ ID NO:4); downstream reverse primer, GTGGCAAAAG CAGCCTTAGC (SEQ ID NO:5); downstream probe, FAM-TCATTCCAGG CCCTTCTTAT TGCATCTG-BHQ (SEQ ID NO:6); in which FAM refers to the 5-carboxyfluorescein fluorescent probe and BHQ refers to the fluorescence quencher of the black hole quencher type (Biosearch Technologies).
  • DNA purified from ES cell clones that have taken up the targeting vector and incorporated in their genomes was combined with TaqManTM Gene Expression Master Mix (Life Technologies) according to the manufacturer's suggestions in a 384-well PCR plate (MicroAmpTM Optical 384-Well Reaction Plate, Life Technologies) and cycled in an Applied Biosystems Prism 7900HT, which collects fluorescence data during the course of the PCRs and determines a threshold cycle (Ct), the fractional PCR cycle at which the accumulated fluorescence reaches a pre-set threshold.
  • Ct threshold cycle
  • the upstream junction of the murine locus and the sequence containing the hIL-6 gene is designed to be within 5'-AATTAGAGAG TTGACTCCTA ATAAATATGA GACTGGGGAT GTCTGTAGCT CATTCTGCTC TGGAGCCCAC CAAGAACGAT AGTCAATTCC AGAAACCGCT ATGAACTCCT TCTCCACAAG TAAGTGCAGG AAATCCTTAG CCCTGGAACT GCCAGCGGCG GTCGAGCCCT GTGTGAGGGA GGGGTGTGTG GCCCAGG (SEQ ID NO:10).
  • the final mouse nucleotide prior to the first nucleotide of the human gene is the "T” in CCGCT
  • the first nucleotide of the human sequence is the first "A” in ATGAA.
  • the downstream junction of the sequence containing the hIL-6 gene and the murine locus is designed to be within 5'-TTTTAAAGAA ATATTTATAT TGTATTTATA TAATGTATAA ATGGTTTTTA TACCAATAAA TGGCATTTTA AAAAATTCAG CAACTTTGAG TGTGTCACGC TCCCGGGCTC GATAACTATA ACGGTCCTAA GGTAGCGACT CGAGATAACT T-3' (SEQ ID NO:11), wherein the final nucleotide of the human sequence is with the final "G” in TCACG and the first nucleotide of the mouse sequence is the first "C” in CTCCC; the downstream junction region also contained a loxP site at the 3' end (the beginning of which is shown) for removal of a floxed
  • the junction of the neo cassette with the mouse IL-6 locus is designed to be within 5'-TATACGAAGT TATCCTAGGT TGGAGCTCCT AAGTTACATC CAAACATCCT CCCCCAAATC AATAATTAAG CACTTTTTAT GACATGTAAA GTTAAATAAG AAGTGAAAGC TGCAGATGGT GAGTGAGA (SEQ ID NO:12), where the final "C” of AGCTC is the final nucleotide of the neo cassette; the first nucleotide of the mouse genome following the cassette is the initial "C" of CTAAG.
  • Humanized IL-6 KI mice were then backcrossed to generate mice lacking Rag 2 and Il2rg and expressing hIL-6, and crossed to mice expressing a human SIRPa transgene ( Strowig et al., 2011, Proc Natl Acad Sci USA, 108(32): 13218-13223 ) to generate mice deficient for Rag2 and Il2rg as well as expressing both hIL-6 and hSIRPa ( Rag2 -/- Il2rg null Il6 h / h hSIRPa + ).
  • mice were crossed with mice expressing human TPO ( Rongvaux et al., 2011, Proc Natl Acad Sci USA, 108(6): 2378-2383 ), human IL-3 and human GM-CSF ( Willinger et al, 2011, Proc Natl Acad Sci USA, 108(6): 2390-2395 ), and human M-CSF ( Rathinam et al, 2011, Blood, 118(11): 3119-3128 ) as well as hSIRPa ( Strowig et al., 2011, Proc Natl Acad Sci USA, 108(32): 13218-13223 ) to generate mice expressing a combination of these human proteins ( Rag2 - / - Il2rg nulll hSIRPa + Tpo h / h Mcsf h / h Il3 / Gmcsf h
  • the multiple myeloma cell line INA-6 ( Burger et al., 2001, Hematol J, 2(1): 42-53 ) was maintained in RPMI1640 medium supplemented with 20 % FCS, penicillin/streptomycin, L-glutamine, and 2.5 ng/ml of hIL-6 in a standard incubator at 37C and 5% CO 2 .
  • mice were irradiated twice with 200 rad from an X-ray source. Indicated amounts of cells were then transplanted into the femur of recipient mice. Briefly, mice were anaesthetized using Ketamine/Xylazine and a hole was drilled into the patellar surface of the femur using a 26 gauge needle. The cells were then slowly injected in a volume of 20 ⁇ l using a 27 gauge needle.
  • mice For transplantation of primary patient-derived cells, Rag2 -/- Il2rg null hSIRPa + Tpo h/h Mcsf h/h Il3/Gmcsf h/h Il6 h/h mice were irradiated twice with 150 rad from an X-ray source and transplantation was performed as described above.
  • ELISA ELISA. Commercial ELISA kits were used to measure the concentrations of human soluble IL-6R (R&D Systems), human Ig ⁇ , and Ig ⁇ (Bethyl Laboratories). Detection of these proteins was performed according to manufacturer's instructions.
  • ⁇ CT Femur morphometry was quantified using cone-beam microfocus x-ray computed tomography ( ⁇ CT40; ScancoMedicalAG). Serial tomographic images were acquired, and 3D images were reconstructed and used to determine the parameters. Trabecular morphometry was characterized by measuring the bone volume fraction, trabecular thickness, trabecular number, and trabecular separation. Cortical measurements included average cortical thickness, cross-sectional area of cortical bone, subperiosteal cross-sectional area, and marrow area.
  • Femurs were stripped of soft tissue, fixed in 10% buffered formalin, dehydrated, and embedded in methyl methacrylate before being sectioned and stained with toluidine blue according to standard procedures.
  • Engraftment of multiple myeloma cell line in mice with humanized IL-6 gene was utilized to evaluate if mice expressing human SIRP ⁇ , and IL-6 are suitable hosts for multiple myeloma (MM) cell lines.
  • the INA6-gfp cell line shows high dependency on human microenvironment, i.e., human fetal bone chips, when transplanted in xenograft systems of scid-hu mice ( Epstein et al., 2005, Methods Mol Med, 113: 183-190 ).
  • INA-6 cells are only able to engraft the human bone graft in scid-hu mice, suggesting dependence on a human bone marrow microenvironment, similar to primary MM cells ( Tassone et al., 2005, Blood, 106(2): 713-716 ).
  • INA-6 cells were transplanted intravenously into i) Rag2 -1- Il2rg null , ii) Rag2 -/- Il2rg null hSIRPa+, iii) Rag2 -/- Il2rg null Il6 h/h , and iv) Rag2 -/- Il2rg null Il6 h/h hSIRPa+ mice. Engraftment was analyzed by measuring sIL-6R protein secreted by INA-6 cells in the blood.
  • INA-6-engrafted mice were treated with Zometa or Velcade, two drugs commonly used to treat multiple myeloma patients. Strikingly, Zometa treatment of mice injected with INA-6 cells was able to reduce bone resorption compared to untreated mice as quantified by ⁇ CT ( Figure 7 ).
  • cytokine genes enable engraftment of primary patient-derived multiple myeloma cells .
  • experiments were conducted to examine the transplantation of primary MM cells into genetically humanized mice. It has been previously demonstrated that humanization of multiple cytokines including thrombopoietin, IL-3, GM-CSF, and M-CSF as well as the macrophage inhibitory receptor SIRPa results in improved engraftment of human hematopoietic cells in immunodeficient mice ( Strowig et al., 2011, Proc Natl Acad Sci USA, 108(32): 13218-13223 ; Rathinam et al, 2011, Blood, 118(11): 3119-3128 ; Rongvaux et al., 2011, Proc Natl Acad Sci USA, 108(6): 2378-2383 ; Willinger et al, 2011, Proc Natl Acad Sci USA, 108(6): 2390-2395 ).
  • IL-3 and GM-CSF improved engraftment of myeloid cells that have been demonstrated to be important for specific aspects of MM pathology.
  • Transgenic expression of hSIRPa improves human cell engraftment, but the SIRPa-CD47 axis has also been recently implicated in tumorgenesis.
  • Previously generated humanized mice were combined with human IL-6 knock-in mice to generate Rug2 - / - Il2rg null hSIRPa + Tpo h / h Mcsf h / h Il3 / Gmcsf h / h Il6 h / h mice.
  • CD3-depleted bone marrow cells from MM patients was injected into the bone marrow of the mice.
  • mice described herein, support the engraftment of primary human MM cells in vivo.
  • the present data demonstrates that the humanization of cytokines gene encoding IL-6, TPO, IL-3, GM-CSF, and/or M-CSF enable engraftment of primary multiple myeloma cells from patients that typically require a human microenvironment for successful transplantation.
  • Example 2 Genetic humanization of the IL-6 gene enables engraftment of mice with human hematopoietic cells.
  • mice The humanized IL-6 KI mouse was generated as described above. The chimeric mice were first bred with BALB/c mice and then backcrossed in order to obtain offspring with hIL-6 in homozygosity. Mice with the same mixed BALB/c x 129 background were used as control.
  • Newborn pups (within first day of life) were sublethally irradiated by X-ray irradiation in a 4-hour interval with 2 x 150cGy.
  • mice Four hours after the irradiation the mice were injected with 1-2x10 5 CD34 + fetal liver (FL) cells in 20 ⁇ l of PBS into the liver by using a 30-gauge needle (Hamilton Company, NV, USA). Mice were weaned at 3 weeks of age. The mice were maintained under specific pathogen-free conditions and all experiments were performed in compliance with Yale Institutional Animal Care and Use Committee protocols.
  • mice were bled from retro-orbital plexus under isofluorane anesthesia at different times after transplantation. Plasma samples were collected and stored at -20°C for further Ig measurement. Red blood cells were lysed two times by using Ammonium-Chloride (ACK) lysing buffer and the remaining PBMC were resuspended in FACS buffer (PBS supplemented with 5%FBS and 5mM EDTA).
  • ACK Ammonium-Chloride
  • mice When the mice were killed, a single-cell suspension of cells was obtained from the bone marrow (BM), thymus and spleen. Samples were then stained with fluorochrome-labeled mAbs against mouse and human cell surface antigens according to the manufactures' instructions. The following anti-human mAbs were used: CD45 (HI30), CD 19 (HIB19), CD3 (UCHT1), CD4 (RPA-T4), CD8 (HIT8a), CD20 (2H7), CD5 (UCHT2), CD27 (O323).
  • CD24 (ML5), CD10 (HI10a), all from Biolegend, CA, USA; CD33 (WM53), CD38 (HIT2), IgM (G20-127), CD138 (M115) from BD Biosciences and CD34 (AC 136) from Miltenyi Biotec.
  • the mouse cells were stained with an anti-murine CD45 Ab (30-F11, Biolegend). Samples were acquired on the LSRII (BD Biosciences) cytometer and analyzed on FlowJo (Treestar, OR, USA) software.
  • Igs Total immunoglobulins
  • 96 well plates (Nunc, NY, USA) were coated at 4°C overnight with 20 ⁇ g/ml purified goat anti-human IgM and IgG (Southern Biotechnology, AL, USA). After washing and blocking with PBS 1% bovine serum albumin (BSA, Sigma-Aldrich) appropriate dilutions of the samples were added for 2 hours at room temperature (RT). Plates were washed and incubated for 1 hour at RT with isotype specific secondary biotinylated antibodies (Southern Biotechnology) followed by Streptavidin-HRP (Pierce Protein Research Products, IL, USA). After a final wash the enzyme activity was determined using the TMB substrate solution followed by the stop solution (both from Pierce Protein Research Products). The absorbance was measured at 450nm. A sample of human serum from Bethyl (TX, USA) was used as reference.
  • B and T cell percentages were similar at week 8 and week 16-20 whereas at weeks 11-15 there was a higher percentage of B cells (69.23 ⁇ 3.97 in IL6m/m and 55.91 ⁇ 4.86 in IL6h/h) than T cells (16.86 ⁇ 3.14 in IL6m/m and 30.26 ⁇ 6.23 in IL6h/h).
  • Myeloid CD33 + cells represented a minor component of the human cell and their percentage decreased over time.
  • the IL6h/h mice showed a sharp increase in the percentage of the CD27 + B cells in the spleen when compared to the IL6m/m mice (24.73% ⁇ 8.94 versus 9.46% ⁇ 2.32) but very few CD27 + cells were found in the BM ( Figure 14A and 14B ).

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Zoology (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Environmental Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Veterinary Medicine (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Husbandry (AREA)
  • Biodiversity & Conservation Biology (AREA)
  • Biomedical Technology (AREA)
  • Immunology (AREA)
  • Urology & Nephrology (AREA)
  • Endocrinology (AREA)
  • Epidemiology (AREA)
  • Diabetes (AREA)
  • Pathology (AREA)
  • Rheumatology (AREA)
  • Public Health (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Cell Biology (AREA)
  • Physics & Mathematics (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
EP17184813.8A 2012-11-05 2013-11-05 Immunodeficient, genetically modified rodent and methods of use thereof Active EP3308641B1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
DK19176252.5T DK3556206T3 (da) 2012-11-05 2013-11-05 Genmodificerede ikke-humane dyr og fremgangsmåder til anvendelse deraf
EP19176252.5A EP3556206B1 (en) 2012-11-05 2013-11-05 Genetically modified non-human animals and methods of use thereof
EP21176673.8A EP3939423B1 (en) 2012-11-05 2013-11-05 Genetically modified non-human animals and methods of use thereof

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201261722437P 2012-11-05 2012-11-05
PCT/US2013/068569 WO2014071397A2 (en) 2012-11-05 2013-11-05 Genetically modified non-human animals and methods of use thereof
EP13811637.1A EP2914102B1 (en) 2012-11-05 2013-11-05 Genetically modified non-human animals and methods of use thereof

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
EP13811637.1A Division EP2914102B1 (en) 2012-11-05 2013-11-05 Genetically modified non-human animals and methods of use thereof
EP13811637.1A Division-Into EP2914102B1 (en) 2012-11-05 2013-11-05 Genetically modified non-human animals and methods of use thereof

Related Child Applications (3)

Application Number Title Priority Date Filing Date
EP19176252.5A Division EP3556206B1 (en) 2012-11-05 2013-11-05 Genetically modified non-human animals and methods of use thereof
EP19176252.5A Division-Into EP3556206B1 (en) 2012-11-05 2013-11-05 Genetically modified non-human animals and methods of use thereof
EP21176673.8A Division EP3939423B1 (en) 2012-11-05 2013-11-05 Genetically modified non-human animals and methods of use thereof

Publications (2)

Publication Number Publication Date
EP3308641A1 EP3308641A1 (en) 2018-04-18
EP3308641B1 true EP3308641B1 (en) 2019-07-03

Family

ID=49876957

Family Applications (4)

Application Number Title Priority Date Filing Date
EP19176252.5A Active EP3556206B1 (en) 2012-11-05 2013-11-05 Genetically modified non-human animals and methods of use thereof
EP21176673.8A Active EP3939423B1 (en) 2012-11-05 2013-11-05 Genetically modified non-human animals and methods of use thereof
EP17184813.8A Active EP3308641B1 (en) 2012-11-05 2013-11-05 Immunodeficient, genetically modified rodent and methods of use thereof
EP13811637.1A Active EP2914102B1 (en) 2012-11-05 2013-11-05 Genetically modified non-human animals and methods of use thereof

Family Applications Before (2)

Application Number Title Priority Date Filing Date
EP19176252.5A Active EP3556206B1 (en) 2012-11-05 2013-11-05 Genetically modified non-human animals and methods of use thereof
EP21176673.8A Active EP3939423B1 (en) 2012-11-05 2013-11-05 Genetically modified non-human animals and methods of use thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
EP13811637.1A Active EP2914102B1 (en) 2012-11-05 2013-11-05 Genetically modified non-human animals and methods of use thereof

Country Status (20)

Country Link
US (5) US9901082B2 (ko)
EP (4) EP3556206B1 (ko)
JP (4) JP6422441B2 (ko)
KR (5) KR102402806B1 (ko)
CN (3) CN104918483B (ko)
AU (3) AU2013337242B2 (ko)
CA (1) CA2887706C (ko)
DK (4) DK2914102T3 (ko)
ES (3) ES2884598T3 (ko)
FI (1) FI3939423T3 (ko)
HK (2) HK1210380A1 (ko)
IL (5) IL311744A (ko)
MX (3) MX370856B (ko)
MY (1) MY175400A (ko)
NO (1) NO2946162T3 (ko)
NZ (3) NZ749626A (ko)
PT (3) PT2914102T (ko)
RU (1) RU2673156C2 (ko)
SG (2) SG11201502685XA (ko)
WO (1) WO2014071397A2 (ko)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11242404B2 (en) 2016-09-21 2022-02-08 ALX Oncology Inc. Antibodies against signal-regulatory protein alpha and methods of use
US11292850B2 (en) 2018-03-21 2022-04-05 ALX Oncology Inc. Antibodies against signal-regulatory protein α and methods of use

Families Citing this family (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050144655A1 (en) 2000-10-31 2005-06-30 Economides Aris N. Methods of modifying eukaryotic cells
US6596541B2 (en) 2000-10-31 2003-07-22 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
AU2010303737B2 (en) 2009-10-06 2014-05-29 Institute For Research In Biomedicine (Irb) Genetically modified mice and engraftment
SG192606A1 (en) 2011-02-15 2013-09-30 Regeneron Pharma Humanized m-csf mice
CN104955326B (zh) 2012-09-07 2018-07-20 再生元制药公司 经遗传修饰的非人动物及其使用方法
CN104918483B (zh) 2012-11-05 2018-05-11 再生元制药公司 生成及使用经遗传修饰的非人动物的方法
HUE041878T2 (hu) 2013-02-22 2019-06-28 Regeneron Pharma Humanizált fõ hisztokompatibilitási komplexet expresszáló egerek
CN105592695B (zh) 2013-09-23 2018-04-10 瑞泽恩制药公司 具有人源化信号调节蛋白基因的非人动物
DK2908626T3 (en) 2013-10-15 2017-03-20 Regeneron Pharma Humanized IL-15 animals
CN106163273B (zh) 2014-04-08 2020-07-24 瑞泽恩制药公司 具有人源化FC-γ受体的非人动物
EP3636073B1 (en) 2014-05-05 2023-11-15 Regeneron Pharmaceuticals, Inc. Humanized c5 and c3 animals
NO2785538T3 (ko) 2014-05-07 2018-08-04
KR20240017405A (ko) * 2014-05-19 2024-02-07 리제너론 파마슈티칼스 인코포레이티드 인간 epo를 발현하는 유전자 변형된 비-인간 동물
RU2735958C2 (ru) 2014-06-19 2020-11-11 Регенерон Фармасьютикалз, Инк. Животные, отличные от человека, имеющие гуманизированный ген 1 запрограммированной гибели клеток
RU2726446C2 (ru) 2014-11-24 2020-07-14 Регенерон Фармасьютикалз, Инк. Не относящиеся к человеку животные, экспрессирующие гуманизированный комплекс cd3
AU2015355328B2 (en) * 2014-12-05 2022-03-10 Regeneron Pharmaceuticals, Inc. Non-human animals having a humanized cluster of differentiation 47 gene
WO2016137841A1 (en) * 2015-02-26 2016-09-01 William Paterson University Of New Jersey Transgenic mice
KR20170133498A (ko) 2015-04-06 2017-12-05 리제너론 파아마슈티컬스, 인크. 비인간 동물에서의 인간화 t 세포 매개 면역반응
KR102658190B1 (ko) 2015-04-13 2024-04-17 리제너론 파마슈티칼스 인코포레이티드 인간화된 sirpa-il15 녹인 마우스 및 이의 이용 방법
PT3376857T (pt) 2015-11-20 2021-05-27 Regeneron Pharma Animais não humanos com um gene humanizado 3 de ativação de linfócitos
JP6980674B2 (ja) 2016-02-29 2021-12-15 リジェネロン・ファーマシューティカルズ・インコーポレイテッドRegeneron Pharmaceuticals, Inc. ヒト化tmprss遺伝子を有する齧歯類
CN109843047B (zh) * 2016-08-11 2022-07-08 杰克逊实验室 涉及经遗传修饰的免疫缺陷型非人类动物中改善的人类红血细胞存活的方法和组合物
CN110740641A (zh) * 2016-11-30 2020-01-31 杰克逊实验室 具有改善的人先天免疫细胞发育的人源化小鼠模型
RU2019125818A (ru) * 2017-02-27 2021-03-29 Регенерон Фармасьютикалс, Инк. Модели ретиношизиса на животных, отличных от человека
CN107115424B (zh) * 2017-05-09 2018-05-29 江西樟树市正康医药生物科技有限公司 改变酸性体质保钙及清热解毒的中药组合物及制备方法
CA3063368A1 (en) * 2017-05-12 2018-11-15 The Jackson Laboratory Nsg mice lacking mhc class i and class ii
US11712026B2 (en) 2017-10-18 2023-08-01 The Jackson Laboratory Murine-MHC-deficient HLA-transgenic nod-mouse models for T1D therapy development
CN116064611A (zh) 2017-11-30 2023-05-05 瑞泽恩制药公司 包含人源化trkb基因座的非人动物
US11576984B2 (en) 2018-03-26 2023-02-14 Regeneron Pharmaceuticals, Inc. Genetically modified mouse with humanized immunoglobulin heavy chain constant region genes and method of using
CN112638154B (zh) 2018-07-16 2022-10-18 瑞泽恩制药公司 Ditra疾病的非人动物模型及其用途
CN111057721B (zh) * 2018-10-12 2021-03-16 百奥赛图(北京)医药科技股份有限公司 人源化IL-4和/或IL-4Rα改造动物模型的制备方法及应用
CN111172190B (zh) * 2018-12-25 2021-07-30 百奥赛图江苏基因生物技术有限公司 人源化细胞因子csf2基因改造非人动物的构建方法及应用
CN114206108B (zh) 2019-04-04 2023-09-29 瑞泽恩制药公司 包括人源化凝血因子12基因座的非人动物
WO2020247452A1 (en) 2019-06-04 2020-12-10 Regeneron Pharmaceuticals, Inc. Non-human animals comprising a humanized ttr locus with a beta-slip mutation and methods of use
EP3796776A1 (en) 2019-06-07 2021-03-31 Regeneron Pharmaceuticals, Inc. Non-human animals comprising a humanized albumin locus
CN111304246B (zh) * 2019-12-17 2021-05-04 百奥赛图江苏基因生物技术有限公司 一种人源化细胞因子动物模型、制备方法及应用
EP4087392A4 (en) * 2020-01-10 2024-02-21 Biocytogen Pharmaceuticals Beijing Co Ltd GENETICALLY MODIFIED NON-HUMAN ANIMAL WITH HUMAN OR CHIMERIC PROTEIN-MHC COMPLEX
US10973908B1 (en) 2020-05-14 2021-04-13 David Gordon Bermudes Expression of SARS-CoV-2 spike protein receptor binding domain in attenuated salmonella as a vaccine
WO2024020057A1 (en) 2022-07-19 2024-01-25 Regeneron Pharmaceuticals, Inc. Genetically modified animal model and its use to model the human immune system

Family Cites Families (72)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4870009A (en) 1982-11-22 1989-09-26 The Salk Institute For Biological Studies Method of obtaining gene product through the generation of transgenic animals
US4736866A (en) 1984-06-22 1988-04-12 President And Fellows Of Harvard College Transgenic non-human mammals
US5573930A (en) 1985-02-05 1996-11-12 Cetus Oncology Corporation DNA encoding various forms of colony stimulating factor-1
JP2644794B2 (ja) 1986-10-24 1997-08-25 カイロン コーポレイション 新規な型のコロニー刺激因子―1
ATE119197T1 (de) 1987-12-23 1995-03-15 Univ Leland Stanford Junior Schimäre immunkompromittierende säugetiere und ihre verwendung.
JP2981486B2 (ja) 1988-06-14 1999-11-22 メディカル・バイオロジー・インスティチュート 哺乳動物の免疫系研究方法
JP2935511B2 (ja) * 1989-10-20 1999-08-16 忠三 岸本 ヒトil―6レセプターの製造方法
US5849288A (en) 1990-01-15 1998-12-15 Yeda Research And Development Co. Ltd. Method for production of monoclonal antibodies in chimeric mice or rats having xenogeneic antibody-producing cells
US5652373A (en) 1990-01-15 1997-07-29 Yeda Research And Development Co. Ltd. Engraftment and development of xenogeneic cells in normal mammals having reconstituted hematopoetic deficient immune systems
DK0438053T3 (da) 1990-01-15 1999-11-22 Yeda Res & Dev Varig transplantation og udvikling af humane hæmopoietiske cellelinjer i normale pattedyr
ATE140480T1 (de) 1990-05-03 1996-08-15 Systemix Inc Menschliches lymph-gewebe in einem wirt mit angegriffenem immunsystem
US5633426A (en) 1990-05-25 1997-05-27 Systemix, Inc. In vivo use of human bone marrow for investigation and production
WO1991018615A1 (en) 1990-05-25 1991-12-12 Systemix, Inc. Human peripheral blood cells in an immunocompromised host
US5222982A (en) 1991-02-11 1993-06-29 Ommaya Ayub K Spinal fluid driven artificial organ
CA2103705C (en) 1991-02-11 1996-01-30 Ayub K. Ommaya Spinal fluid driven artificial organ
EP0517199A1 (en) 1991-06-04 1992-12-09 Yeda Research And Development Company, Ltd. Durable engraftment of human tissue and cells in normal mammals
WO1993005796A1 (en) 1991-09-19 1993-04-01 The Scripps Research Institute Method for producing human antibodies in a non-human animal, and animals therefor
EP0539970B1 (en) 1991-10-30 1999-05-26 Idemitsu Kosan Company Limited Methods for producing human lymphocytes and human monoclonal antibodies, and human monoclonal antibodies produced thereby
US6353150B1 (en) 1991-11-22 2002-03-05 Hsc Research And Development Limited Partnership Chimeric mammals with human hematopoietic cells
WO1993018144A1 (en) 1992-03-05 1993-09-16 The Trustees Of Columbia University Of The City Of New York Recombination activating gene deficient animal
US5866757A (en) 1992-06-02 1999-02-02 Yeda Research And Development Co. Ltd. Engraftment and development of xenogeneic cells in normal mammals having reconstituted hematopoetic deficient immune systems
US6018096A (en) 1993-05-03 2000-01-25 Surrogen, Inc. Animal model for engraftment, proliferation and differentiation of human hematopoietic stem cells
CA2103693A1 (en) 1993-08-06 1995-02-07 Steven Gallinger Animal model of the human immune system
EP0771348A1 (en) 1994-07-27 1997-05-07 Merck & Co. Inc. Bradykinin b2 receptor modified transgenic non-human animals
US6455756B1 (en) 1994-08-12 2002-09-24 Novartis Ag Long term xenogeneic myeloid and lymphoid cell production in chimeric immunocompromised mice
US7273753B2 (en) 1996-08-02 2007-09-25 Center Of Blood Research Purification and uses of dendritic cells and monocytes
JP4312265B2 (ja) 1997-04-09 2009-08-12 ラング−ジ チャン ワクチン評価のための動物モデル
US6248721B1 (en) 1997-04-09 2001-06-19 Lung-Ji Chang Method of using mouse model for evaluation of HIV vaccines
GB2337755B (en) * 1998-05-29 2003-10-29 Secr Defence Virus vaccine
AU7340000A (en) 1999-08-31 2001-03-26 Genencor International, Inc. Transgenic mammal capable of facilitating production of donor-specific functional immunity
US20030028911A1 (en) 1999-08-31 2003-02-06 Manley Huang Transgenic mammal capable of facilitating production of donor-specific functional immunity
EP1244807A1 (en) 1999-12-09 2002-10-02 Human Genome Sciences, Inc. Il-6 like polynucleotide
WO2002022786A1 (en) 2000-09-14 2002-03-21 Genetrol Biotherapeutics, Inc. Method and cell composition for screening compounds for anti-inflammatory activity
US6586251B2 (en) 2000-10-31 2003-07-01 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
US6596541B2 (en) 2000-10-31 2003-07-22 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
CA2453531A1 (en) * 2001-07-10 2003-01-23 Johnson & Johnson Research Pty Limited Methods for genetic modification of hematopoietic progenitor cells and uses of the modified cells
WO2003039232A2 (en) 2001-10-26 2003-05-15 Large Scale Biology Corporation Endothelial cell derived hemotopoietic growth factor
JP4087338B2 (ja) 2001-11-15 2008-05-21 キリンファーマ株式会社 キメラ非ヒト動物
JPWO2004005496A1 (ja) 2002-07-05 2005-11-04 麒麟麦酒株式会社 臍帯血、骨髄、末梢血等に含まれる新規な未分化幹細胞集団
CN101250553A (zh) 2002-07-13 2008-08-27 上海医学遗传研究所 一种促人血小板生成素表达载体及其构建方法
US20040128703A1 (en) 2002-09-09 2004-07-01 Hiroaki Shizuya Methods and compositions for the generation of humanized mice
EP1418185A1 (en) 2002-11-11 2004-05-12 Aventis Pharma Deutschland GmbH Use of EDG2 receptor in an animal model of heart failure
US7402728B2 (en) 2002-12-16 2008-07-22 Genentech, Inc. Transgenic mice expressing human CD20 and/or CD16
WO2004081229A2 (en) * 2003-03-12 2004-09-23 Genentech, Inc. Use of bv8 and/or eg-vegf to promote hematopoiesis
US20080311095A1 (en) 2007-05-23 2008-12-18 Sangamo Biosciences, Inc. Methods and compositions for increased transgene expression
EP1598364A1 (en) * 2004-05-21 2005-11-23 AGIRx Limited Chimerical soluble hyper IL-11 receptor and use thereof
DK2767161T3 (en) 2004-10-19 2018-05-07 Regeneron Pharma Method of generating an animal homozygous for genetic modification
US7759541B2 (en) * 2004-12-13 2010-07-20 Iti Life Sciences Transgenic animals for assessing drug metabolism and toxicity
GB2434578A (en) 2006-01-26 2007-08-01 Univ Basel Transgenic animals
ES2618787T5 (es) 2006-04-25 2022-10-21 Univ California Administración de factores de crecimiento para el tratamiento de trastornos del SNC
EP1878342A1 (en) 2006-07-13 2008-01-16 Institut Pasteur Immunodeficient mice transgenic for HLA class I and HLA class II molecules and their uses
WO2008060360A2 (en) 2006-09-28 2008-05-22 Surmodics, Inc. Implantable medical device with apertures for delivery of bioactive agents
WO2008069659A1 (en) 2006-12-05 2008-06-12 Academisch Ziekenhuis Bij De Universiteit Van Amsterdam Improved xenogenic immune system in a non-human mammal
GB0718029D0 (en) 2007-09-14 2007-10-24 Iti Scotland Ltd Two step cluster deletion and humanisation
WO2009042917A1 (en) 2007-09-28 2009-04-02 The General Hospital Corporation Methods and compositions for antibody production
WO2009097468A2 (en) 2008-01-29 2009-08-06 Kliman Gilbert H Drug delivery devices, kits and methods therefor
BRPI1013771A2 (pt) * 2009-04-13 2016-04-05 Apceth Gmbh & Co Kg "células-tronco mesenquimais projetadas e método de uso das mesmas para tratar tumores."
DK2448967T3 (en) 2009-06-29 2015-07-20 Ilya B Leskov NON-HUMAN MAMMALS MODEL OF CANCER human hematopoietic
JP2012531896A (ja) * 2009-06-29 2012-12-13 チェン,チンフェン ヒト化非ヒト哺乳動物を製造する方法
RU2425880C2 (ru) 2009-07-30 2011-08-10 Учреждение Российской академии наук Институт общей генетики им. Н.И. Вавилова РАН Способ получения трансгенных мышей
AU2010303737B2 (en) 2009-10-06 2014-05-29 Institute For Research In Biomedicine (Irb) Genetically modified mice and engraftment
WO2011084664A1 (en) * 2009-12-21 2011-07-14 Regeneron Pharmaceuticals, Inc. HUMANIZED FCγ R MICE
EP2618656B1 (en) * 2010-09-20 2018-06-20 Yale University, Inc. HUMAN SIRPalpha TRANSGENIC ANIMALS AND THEIR METHODS OF USE
WO2012051572A1 (en) 2010-10-15 2012-04-19 Massachusetts Institute Of Technology A humanized non-human mammal model of malaria and uses thereof
SG192606A1 (en) 2011-02-15 2013-09-30 Regeneron Pharma Humanized m-csf mice
DK2818478T3 (en) * 2011-10-28 2017-05-22 Regeneron Pharma HUMANIZED IL-6 AND IL-6 RECEPTOR
CN104955326B (zh) * 2012-09-07 2018-07-20 再生元制药公司 经遗传修饰的非人动物及其使用方法
CN104918483B (zh) 2012-11-05 2018-05-11 再生元制药公司 生成及使用经遗传修饰的非人动物的方法
CN105592695B (zh) 2013-09-23 2018-04-10 瑞泽恩制药公司 具有人源化信号调节蛋白基因的非人动物
DK2908626T3 (en) 2013-10-15 2017-03-20 Regeneron Pharma Humanized IL-15 animals
KR20240017405A (ko) 2014-05-19 2024-02-07 리제너론 파마슈티칼스 인코포레이티드 인간 epo를 발현하는 유전자 변형된 비-인간 동물
KR102658190B1 (ko) 2015-04-13 2024-04-17 리제너론 파마슈티칼스 인코포레이티드 인간화된 sirpa-il15 녹인 마우스 및 이의 이용 방법

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11242404B2 (en) 2016-09-21 2022-02-08 ALX Oncology Inc. Antibodies against signal-regulatory protein alpha and methods of use
US11401338B2 (en) 2016-09-21 2022-08-02 ALX Oncology Inc. Antibodies against signal-regulatory protein alpha and methods of use
US11292850B2 (en) 2018-03-21 2022-04-05 ALX Oncology Inc. Antibodies against signal-regulatory protein α and methods of use
US11939393B2 (en) 2018-03-21 2024-03-26 ALX Oncology Inc. Antibodies against signal-regulatory protein alpha and methods of use

Also Published As

Publication number Publication date
KR102275412B1 (ko) 2021-07-12
US20240099278A1 (en) 2024-03-28
PT3308641T (pt) 2019-10-24
JP6772234B2 (ja) 2020-10-21
KR102402806B1 (ko) 2022-05-30
MX2020012427A (es) 2022-06-08
ES2655442T3 (es) 2018-02-20
US9986724B2 (en) 2018-06-05
US11778995B2 (en) 2023-10-10
MX2019015882A (es) 2020-02-07
DK3556206T3 (da) 2021-08-09
IL299887A (en) 2023-03-01
AU2013337242B2 (en) 2019-04-04
CN108401987B (zh) 2022-04-29
ES2748662T3 (es) 2020-03-17
IL258968B (en) 2019-10-31
IL299887B1 (en) 2024-05-01
CN104918483A (zh) 2015-09-16
EP3556206B1 (en) 2021-06-02
WO2014071397A3 (en) 2014-06-26
EP3308641A1 (en) 2018-04-18
AU2013337242A1 (en) 2015-04-30
SG11201502685XA (en) 2015-05-28
IL238478B (en) 2018-05-31
KR20210087567A (ko) 2021-07-12
JP2015534819A (ja) 2015-12-07
KR20150081285A (ko) 2015-07-13
NO2946162T3 (ko) 2018-06-09
ES2884598T3 (es) 2021-12-10
RU2673156C2 (ru) 2018-11-22
RU2018140189A (ru) 2018-12-10
JP6422441B2 (ja) 2018-11-14
AU2022200302A1 (en) 2022-02-10
RU2015121195A (ru) 2016-12-27
KR20240042142A (ko) 2024-04-01
KR102649893B1 (ko) 2024-03-25
MX2015005654A (es) 2016-04-28
US20140134662A1 (en) 2014-05-15
NZ724353A (en) 2022-05-27
JP7105844B2 (ja) 2022-07-25
IL270124B1 (ko) 2023-02-01
US10785968B2 (en) 2020-09-29
JP2022132376A (ja) 2022-09-08
CN104918483B (zh) 2018-05-11
IL270124B2 (en) 2023-06-01
US9901082B2 (en) 2018-02-27
KR20220148307A (ko) 2022-11-04
CN108401987A (zh) 2018-08-17
RU2018140189A3 (ko) 2022-05-04
EP3556206A1 (en) 2019-10-23
EP2914102A2 (en) 2015-09-09
CN114766432A (zh) 2022-07-22
NZ631614A (en) 2016-10-28
KR102458016B1 (ko) 2022-10-25
DK3308641T3 (da) 2019-10-07
AU2019204775A1 (en) 2019-07-25
MY175400A (en) 2020-06-24
HK1253909A1 (zh) 2019-07-05
PT2914102T (pt) 2018-01-15
EP3939423A1 (en) 2022-01-19
US20180049413A1 (en) 2018-02-22
JP2021010371A (ja) 2021-02-04
HK1210380A1 (en) 2016-04-22
IL311744A (en) 2024-05-01
SG10201707449TA (en) 2017-10-30
US20160366862A1 (en) 2016-12-22
US20210100228A1 (en) 2021-04-08
DK2914102T3 (en) 2017-12-11
FI3939423T3 (fi) 2024-05-23
MX370856B (es) 2020-01-08
IL270124A (ko) 2019-12-31
WO2014071397A2 (en) 2014-05-08
JP2019037238A (ja) 2019-03-14
IL238478A0 (en) 2015-06-30
PT3556206T (pt) 2021-08-24
DK3939423T3 (da) 2024-05-06
EP3939423B1 (en) 2024-03-06
KR20220075242A (ko) 2022-06-07
AU2019204775B2 (en) 2021-10-21
CA2887706A1 (en) 2014-05-08
NZ749626A (en) 2023-11-24
CA2887706C (en) 2022-08-02
EP2914102B1 (en) 2017-10-18
IL258968A (en) 2018-06-28

Similar Documents

Publication Publication Date Title
US11778995B2 (en) Genetically modified non-human animals and methods of use thereof
RU2775425C2 (ru) Генетически модифицированные не относящиеся к человеческому роду животные и способы их применения

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN PUBLISHED

AC Divisional application: reference to earlier application

Ref document number: 2914102

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20181017

RBV Designated contracting states (corrected)

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20190116

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

AC Divisional application: reference to earlier application

Ref document number: 2914102

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1253909

Country of ref document: HK

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

Ref country code: AT

Ref legal event code: REF

Ref document number: 1149867

Country of ref document: AT

Kind code of ref document: T

Effective date: 20190715

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602013057548

Country of ref document: DE

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

Effective date: 20191003

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: NL

Ref legal event code: FP

REG Reference to a national code

Ref country code: PT

Ref legal event code: SC4A

Ref document number: 3308641

Country of ref document: PT

Date of ref document: 20191024

Kind code of ref document: T

Free format text: AVAILABILITY OF NATIONAL TRANSLATION

Effective date: 20191001

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

Ref country code: NO

Ref legal event code: T2

Effective date: 20190703

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190703

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190703

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20191003

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: AL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190703

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190703

Ref country code: RS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190703

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20191004

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20191103

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2748662

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20200317

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190703

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190703

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190703

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190703

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190703

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200224

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190703

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602013057548

Country of ref document: DE

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

PG2D Information on lapse in contracting state deleted

Ref country code: IS

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190703

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20191105

26N No opposition filed

Effective date: 20200603

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190703

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190703

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20131105

Ref country code: MT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190703

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190703

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230513

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: NL

Payment date: 20231020

Year of fee payment: 11

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20231019

Year of fee payment: 11

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: ES

Payment date: 20231201

Year of fee payment: 11

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: SE

Payment date: 20231020

Year of fee payment: 11

Ref country code: PT

Payment date: 20231019

Year of fee payment: 11

Ref country code: NO

Payment date: 20231023

Year of fee payment: 11

Ref country code: IT

Payment date: 20231019

Year of fee payment: 11

Ref country code: IE

Payment date: 20231023

Year of fee payment: 11

Ref country code: FR

Payment date: 20231019

Year of fee payment: 11

Ref country code: FI

Payment date: 20231019

Year of fee payment: 11

Ref country code: DK

Payment date: 20231019

Year of fee payment: 11

Ref country code: DE

Payment date: 20231019

Year of fee payment: 11

Ref country code: CZ

Payment date: 20231023

Year of fee payment: 11

Ref country code: CH

Payment date: 20231201

Year of fee payment: 11

Ref country code: AT

Payment date: 20231023

Year of fee payment: 11

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: BE

Payment date: 20231019

Year of fee payment: 11